1
|
Wei L, Zhu W, Dong C, Kim JK, Ma Y, Denning TL, Kang SM, Wang BZ. Lipid nanoparticles encapsulating both adjuvant and antigen mRNA improve influenza immune cross-protection in mice. Biomaterials 2025; 317:123039. [PMID: 39724768 DOI: 10.1016/j.biomaterials.2024.123039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/26/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
The rapid approval of SARS-CoV-2 mRNA lipid nanoparticle (LNP) vaccines indicates the versatility of mRNA LNPs in an urgent vaccine need. However, the mRNA vaccines do not induce mucosal cellular responses or broad protection against recent variants. To improve cross-protection of mRNA vaccines, here we engineered a pioneered mRNA LNP encapsulating with mRNA constructs encoding cytokine adjuvant and influenza A hemagglutinin (HA) antigen for intradermal vaccination. The adjuvant mRNA encodes a novel fusion cytokine GIFT4 comprising GM-CSF and IL-4. We found that the adjuvanted mRNA LNP vaccine induced high levels of humoral antibodies and systemic T cell responses against heterologous influenza antigens and protected immunized mice against influenza A viral infections. Also, the adjuvanted mRNA LNP vaccine elicited early germinal center reactions in draining lymph nodes and promoted antibody-secreting B cell responses. In addition, we generated another adjuvant mRNA encoding CCL27, which enhanced systemic immune responses. We found the two adjuvant mRNAs both showed effective adjuvanticity in enhancing humoral and cellular responses in mice. Interestingly, intradermal immunizations of GIFT4 or CCL27 mRNA adjuvanted mRNA LNP vaccines induced significant lung tissue-resident T cells. Our findings demonstrate that the cytokine mRNA can be a promising adjuvant flexibly formulated into mRNA LNP vaccines to provoke strong immunity against viral variants.
Collapse
Affiliation(s)
- Lai Wei
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Liaw K, Konrath KM, Trachtman AR, Tursi NJ, Gary EN, Livingston C, Flowers K, Chu JD, Hojecki CE, Laenger N, McCanna ME, Agostino CJ, Chokkalingam N, Bayruns K, Kriete S, Kim A, Park J, Monastra C, Pardo LA, Jenison S, Huang J, Mulka K, Patel A, Kulp DW, Weiner DB. DNA co-delivery of seasonal H1 influenza hemagglutinin nanoparticle vaccines with chemokine adjuvant CTACK induces potent immunogenicity for heterologous protection in vivo. Vaccine 2025; 59:127231. [PMID: 40398322 DOI: 10.1016/j.vaccine.2025.127231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 04/08/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
Current influenza vaccines induce mostly strain-specific immunity necessitating annual reformulation and dosing. Here, we developed an improved seasonal influenza vaccine based on A/H1N1/Wisconsin/588/2019. We designed a DNA-launched self-assembling nanoparticle that displayed seven Wisconsin/588/2019 hemagglutinin (HA) head domains (WI19-7mer). WI19-7mer nanovaccine improved heterologous HAI titers and CD8+ cellular responses in mice than DNA encoded HA trimer (WI19 HA). In human antibody repertoire mice, WI19-7mer induced superior breadth to a diverse panel of H1 HAs compared to WI19 HA immunized animals. Cross-reactive HAI titers were maintained better in mice immunized with WI19-7mer than WI19 HA. The WI19-7mer induced improved antibody binding breadth and provided superior protection in a heterologous challenge compared to challenge-matched HA trimer. Addition of the cytokine adjuvant (CTACK) to WI19-7mer significantly improved breadth, HAI, peripheral responses, and protection in heterologous challenge. These data demonstrate that combining nucleic acid delivery, immune focusing, low valency nanoparticle, and mucosal adjuvant for enhanced vaccine effectiveness has broader applications for other viruses.
Collapse
Affiliation(s)
- Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Kylie M Konrath
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Abigail R Trachtman
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ebony N Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Cory Livingston
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Kaitlyn Flowers
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacqueline D Chu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Casey E Hojecki
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Niklas Laenger
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Biology Department, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Madison E McCanna
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Colby J Agostino
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neethu Chokkalingam
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Kelly Bayruns
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Sinja Kriete
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Amber Kim
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Joyce Park
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Cara Monastra
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Lucas A Pardo
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Sarah Jenison
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jinwei Huang
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Kathleen Mulka
- Penn Vet Comparative Pathology Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Daniel W Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Wang J, Li X, Li C, Liu L, Wang Z, Feng J. The Codonopsis pilosula water extract improves testicular inflammatory aging in D-galactose induced aging mice by modulating the CLEC7A/inflammasome pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119645. [PMID: 40113110 DOI: 10.1016/j.jep.2025.119645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
AIM OF THE STUDY Aging-induced testicular inflammation impairs male fertility. The purpose of this study was to investigate the effectiveness and mechanism of C. pilosula water extract (CPWE) in preventing testicular inflammation in D-galactose-induced aging mice. MATERIALS AND METHODS The "The Plant List" database (www.theplantlist.org) provided verified plant taxonomy. D-galactose was intraperitoneally injected to induce an aging mice model, with high, medium, and low dosages of CPWE used as pharmacological interventions. The concentrations of superoxide dismutase (SOD), malondialdehyde (MDA), testosterone and in mouse serum or testicle samples after CPWE treatment were quantified using biochemical method. Hematoxylin and eosin (HE) staining was employed to assess the morphological features of testicular tissues, whereas immunohistochemical (IHC) analysis and enzyme-linked immunosorbent assay (ELISA) were conducted to evaluate the presence and levels of inflammatory cytokines interleukin-6 (IL-6) and interleukin-1β (IL-1β) within testicular samples of mice. Differentially expressed genes were identified using transcriptome sequencing; the genes and pathways regulated by CPWE, as well as immune cell infiltration, were examined using bioinformatics analysis. The expression of target gene and pathway-related protein was confirmed using real-time quantitative PCR and Western blotting. RESULTS Treatment with CPWE alleviated the pathological alterations in the testicular tissues of aged mice, increased the concentrations of SOD and testosterone in the serum, and decreased the levels of MDA, IL-6 and IL-1β in the testes. The expression of C-C motif chemokine ligand 21a (Ccl21a) and C-C motif chemokine ligand 27b (Ccl27b) genes was downregulated after treatment with CPWE. The protein levels associated with the C-type lectin domain family 7, member A (CLEC7A)/inflammasome signaling pathway, including IL-1β, Caspase 8 (CASP8), and nuclear factor-kappa B (NF-κB), were found to be downregulated after treatment with CPWE. T cells, B cells, and macrophages showed a strong association with aging and the modulatory effects of CPWE. CONCLUSIONS The results indicate that CPWE regulates the CLEC7A/inflammasome pathway, thereby inhibiting inflammasomes activation and reducing the expressions of proinflammatory cytokines such as IL-6 and IL-1β, as well as chemokines such as Ccl21a and Ccl27b, providing substantial protection against age-related testicular inflammatory injury.
Collapse
Affiliation(s)
- Jing Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Xuechan Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Caihong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Lijun Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
| | - Zhenjuan Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, China.
| |
Collapse
|
4
|
Lee MJ, Hammouda MB, Miao W, Okafor AE, Jin YJ, Sun H, Jain V, Markovtsov V, Diao Y, Gregory SG, Zhang JY. UBE2N Is Essential for Maintenance of Skin Homeostasis and Suppression of Inflammation. J Invest Dermatol 2024; 144:2742-2753. [PMID: 38796140 PMCID: PMC11581932 DOI: 10.1016/j.jid.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/28/2024]
Abstract
UBE2N, a Lys63 ubiquitin-conjugating enzyme, plays critical roles in embryogenesis and immune system development and function. However, its roles in adult epithelial tissue homeostasis and pathogenesis are unclear. We generated conditional mouse models that deleted Ube2n in skin cells in a temporally and spatially controlled manner. We found that Ube2n knockout in the adult skin keratinocytes induced a range of inflammatory skin defects characteristic of psoriatic and actinic keratosis. These included inflammation, epidermal and dermal thickening, parakeratosis, and increased immune cell infiltration as well as signs of edema and blistering. Single-cell transcriptomic analyses and RT-qPCR showed that Ube2n-knockout keratinocytes expressed elevated myeloid cell chemoattractants such as Cxcl1 and Cxcl2 and decreased the homeostatic T lymphocyte chemoattractant Ccl27a. Consistently, the infiltrating immune cells were predominantly myeloid-derived cells, including neutrophils and M1-like macrophages, which expressed high levels of inflammatory cytokines such as Il1β and Il24. Pharmacological blockade of the IL-1 receptor associated kinases (IRAK1/4) alleviated inflammation, epidermal and dermal thickening, and immune infiltration of the Ube2n-mutant skin. Together, these findings highlight a key role of keratinocyte UBE2N in maintenance of epidermal homeostasis and skin immunity and identify IRAK1/4 as potential therapeutic target for inflammatory skin disorders.
Collapse
Affiliation(s)
- Min Jin Lee
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA; Department of Molecular Genetics & Microbiology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Manel Ben Hammouda
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Wanying Miao
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Arinze E Okafor
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Yingai J Jin
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Huiying Sun
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | | | - Yarui Diao
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Jennifer Y Zhang
- Department of Dermatology, School of Medicine, Duke University, Durham, North Carolina, USA; Department of Pathology, School of Medicine, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
5
|
Rantasalo V, Laukka D, Nikulainen V, Jalkanen J, Gunn J, Kiviniemi T, Hakovirta H. Association between aortic calcification and cytokine levels in patients with peripheral artery disease. Clin Transl Sci 2024; 17:e70036. [PMID: 39344403 PMCID: PMC11440032 DOI: 10.1111/cts.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/10/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Aortic calcification-a marker of advanced atherosclerosis in large arteries-associates with cardiovascular mortality and morbidity. Little is known about the soluble inflamJarmatory profiles involved in large artery atherosclerosis. We investigated the correlation between aortic calcification in the abdominal aorta and cytokine levels in a cohort of peripheral artery disease patients. Aortic calcification index was measured from computed tomography exams and circulating cytokine levels were analyzed from blood serum samples of 156 consecutive patients prior to invasive treatment of peripheral artery disease. The study included 156 patients (mean age 70.7 years, 64 (41.0%) women). The mean ankle-brachial index (ABI) was 0.64 and the mean aortic calcification index (ACI) was 52.3. ACI was associated with cytokines cutaneous T-cell-attracting chemokine CTACK (β 23.08, SE 5.22, p < 0.001) and monokine induced by gamma-interferon MIG (β 9.40, SE 2.82, p 0.001) in univariate linear regression. After adjustment with cardiovascular risk factors, CTACK and MIG were independently associated with ACI, β 17.9 (SE 5.22, p < 0.001) for CTACK and β 6.80 (SE 3.33, p 0.043) for MIG. CTACK was significantly higher in the patients representing the highest ACI tertile (highest vs. middle, 7.53 vs. 7.34 Tukeys HSD p-value 0.023 and highest vs. lowest tertile 7.53 vs. 7.29, Tukeys HSD p-value 0.002). MIG was significantly higher in the highest tertile versus lowest (7.65 vs. 7.30, Tukeys HSD p-value 0.027). Cytokines CTACK and MIG are associated with higher ACI, suggesting that CTACK and MIG reflect atherosclerotic disease burden of the aorta. This might further suggest the possible association with other cardiovascular morbidities.
Collapse
Affiliation(s)
- Ville Rantasalo
- Department of Surgery, University of Turku, Turku, Finland
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Dan Laukka
- Department of Clinical Neurosciences, University of Turku, Turku, Finland
- Department of Neurosurgery, Neurocenter, Turku University Hospital, Turku, Finland
| | - Veikko Nikulainen
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Juho Jalkanen
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Jarmo Gunn
- Department of Surgery, University of Turku, Turku, Finland
| | | | - Harri Hakovirta
- Department of Surgery, University of Turku, Turku, Finland
- Satasairaala, Pori, Finland
- King Faisal Specialist Hospital and Research Center, Medina, Saudi Arabia
| |
Collapse
|
6
|
Sun P, Kraus CN, Zhao W, Xu J, Suh S, Nguyen Q, Jia Y, Nair A, Oakes M, Tinoco R, Shiu J, Sun B, Elsensohn A, Atwood SX, Nie Q, Dai X. Single-cell and spatial transcriptomics of vulvar lichen sclerosus reveal multi-compartmental alterations in gene expression and signaling cross-talk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607986. [PMID: 39211101 PMCID: PMC11361165 DOI: 10.1101/2024.08.14.607986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Vulvar diseases are a critical yet often neglected area of women's health, profoundly affecting patients' quality of life and frequently resulting in long-term physical and psychological challenges. Lichen sclerosus (LS) is a chronic inflammatory skin disorder that predominantly affects the vulva, leading to severe itching, pain, scarring, and an increased risk of malignancy. Despite its profound impact on affected individuals, the molecular pathogenesis of vulvar LS (VLS) is not well understood, hindering the development of FDA-approved therapies. Here, we utilize single-cell and spatial transcriptomics to analyze lesional and non-lesional skin from VLS patients, as well as healthy control vulvar skin. Our findings demonstrate histologic, cellular, and molecular heterogeneities within VLS, yet highlight unifying molecular changes across keratinocytes, fibroblasts, immune cells, and melanocytes in lesional skin. They reveal cellular stress and damage in fibroblasts and keratinocytes, enhanced T cell activation and cytotoxicity, aberrant cell-cell signaling, and increased activation of the IFN, JAK/STAT, and p53 pathways in specific cell types. Using both monolayer and organotypic culture models, we also demonstrate that knockdown of select genes, which are downregulated in VLS lesional keratinocytes, partially recapitulates VLS-like stress-associated changes. Collectively, these data provide novel insights into the pathogenesis of VLS, identifying potential biomarkers and therapeutic targets for future research.
Collapse
|
7
|
Elmore AR, Adhikari N, Hartley AE, Aparicio HJ, Posner DC, Hemani G, Tilling K, Gaunt TR, Wilson PW, Casas JP, Gaziano JM, Davey Smith G, Paternoster L, Cho K, Peloso GM. Protein Identification for Stroke Progression via Mendelian Randomization in Million Veteran Program and UK Biobank. Stroke 2024; 55:2045-2054. [PMID: 39038097 PMCID: PMC11259242 DOI: 10.1161/strokeaha.124.047103] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Individuals who have experienced a stroke, or transient ischemic attack, face a heightened risk of future cardiovascular events. Identification of genetic and molecular risk factors for subsequent cardiovascular outcomes may identify effective therapeutic targets to improve prognosis after an incident stroke. METHODS We performed genome-wide association studies for subsequent major adverse cardiovascular events (MACE; ncases=51 929; ncontrols=39 980) and subsequent arterial ischemic stroke (AIS; ncases=45 120; ncontrols=46 789) after the first incident stroke within the Million Veteran Program and UK Biobank. We then used genetic variants associated with proteins (protein quantitative trait loci) to determine the effect of 1463 plasma protein abundances on subsequent MACE using Mendelian randomization. RESULTS Two variants were significantly associated with subsequent cardiovascular events: rs76472767 near gene RNF220 (odds ratio, 0.75 [95% CI, 0.64-0.85]; P=3.69×10-8) with subsequent AIS and rs13294166 near gene LINC01492 (odds ratio, 1.52 [95% CI, 1.37-1.67]; P=3.77×10-8) with subsequent MACE. Using Mendelian randomization, we identified 2 proteins with an effect on subsequent MACE after a stroke: CCL27 ([C-C motif chemokine 27], effect odds ratio, 0.77 [95% CI, 0.66-0.88]; adjusted P=0.05) and TNFRSF14 ([tumor necrosis factor receptor superfamily member 14], effect odds ratio, 1.42 [95% CI, 1.24-1.60]; adjusted P=0.006). These proteins are not associated with incident AIS and are implicated to have a role in inflammation. CONCLUSIONS We found evidence that 2 proteins with little effect on incident stroke appear to influence subsequent MACE after incident AIS. These associations suggest that inflammation is a contributing factor to subsequent MACE outcomes after incident AIS and highlights potential novel targets.
Collapse
Affiliation(s)
- Andrew R. Elmore
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Nimish Adhikari
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
- Department of Biostatistics, Boston University School of Public Health, MA (N.A., G.M.P.)
| | - April E. Hartley
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Hugo Javier Aparicio
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA (H.J.A.)
- Boston Medical Center, MA (H.J.A.)
| | - Daniel C. Posner
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
| | - Gibran Hemani
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Kate Tilling
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Tom R. Gaunt
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | | | - Juan P. Casas
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.P.C., J.M.G., K.C.)
| | - John Michael Gaziano
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.P.C., J.M.G., K.C.)
| | - George Davey Smith
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Lavinia Paternoster
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, United Kingdom (A.R.E., A.E.H., G.H., K.T., T.R.G., G.D.S., L.P.)
| | - Kelly Cho
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.P.C., J.M.G., K.C.)
| | - Gina M. Peloso
- Veteran’s Affairs Healthcare System, Boston, MA (N.A., D.C.P., J.P.C., J.M.G., K.C., G.M.P.)
- Department of Biostatistics, Boston University School of Public Health, MA (N.A., G.M.P.)
| |
Collapse
|
8
|
Song X, Yao Z, Zhang Z, Lyu S, Chen N, Qi X, Liu X, Ma W, Wang W, Lei C, Jiang Y, Wang E, Huang Y. Whole-genome sequencing reveals genomic diversity and selection signatures in Xia'nan cattle. BMC Genomics 2024; 25:559. [PMID: 38840048 PMCID: PMC11151506 DOI: 10.1186/s12864-024-10463-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The crossbreeding of specialized beef cattle breeds with Chinese indigenous cattle is a common method of genetic improvement. Xia'nan cattle, a crossbreed of Charolais and Nanyang cattle, is China's first specialized beef cattle breed with independent intellectual property rights. After more than two decades of selective breeding, Xia'nan cattle exhibit a robust physique, good environmental adaptability, good tolerance to coarse feed, and high meat production rates. This study analyzed the population genetic structure, genetic diversity, and genomic variations of Xia'nan cattle using whole-genome sequencing data from 30 Xia'nan cattle and 178 published cattle genomic data. RESULT The ancestry estimating composition analysis showed that the ancestry proportions for Xia'nan cattle were mainly Charolais with a small amount of Nanyang cattle. Through the genetic diversity studies (nucleotide diversity and linkage disequilibrium decay), we found that the genomic diversity of Xia'nan cattle is higher than that of specialized beef cattle breeds in Europe but lower than that of Chinese native cattle. Then, we used four methods to detect genome candidate regions influencing the excellent traits of Xia'nan cattle. Among the detected results, 42 genes (θπ and CLR) and 131 genes (FST and XP-EHH) were detected by two different detection strategies. In addition, we found a region in BTA8 with strong selection signals. Finally, we conducted functional annotation on the detected genes and found that these genes may influence body development (NR6A1), meat quality traits (MCCC1), growth traits (WSCD1, TMEM68, MFN1, NCKAP5), and immunity (IL11RA, CNTFR, CCL27, SLAMF1, SLAMF7, NAA35, and GOLM1). CONCLUSION We elucidated the genomic features and population structure of Xia'nan cattle and detected some selection signals in genomic regions potentially associated with crucial economic traits in Xia'nan cattle. This research provided a basis for further breeding improvements in Xia'nan cattle and served as a reference for genetic enhancements in other crossbreed cattle.
Collapse
Affiliation(s)
- Xingya Song
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China
| | - Zhi Yao
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China
| | - Zijing Zhang
- Institute of Animal Husbandry, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, Henan, People's Republic of China
| | - Shijie Lyu
- Institute of Animal Husbandry, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, Henan, People's Republic of China
| | - Ningbo Chen
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China
| | - Xingshan Qi
- Biyang County Xiananniu Technology Development Co., Ltd, Zhumadian, 463700, People's Republic of China
| | - Xian Liu
- Henan Provincial Livestock Technology Promotion Station, Zhengzhou, 450008, Henan, People's Republic of China
| | - Weidong Ma
- Shaanxi Agricultural and Animal Husbandry Seed Farm, Shaanxi Fufeng, 722203, People's Republic of China
| | - Wusheng Wang
- Shaanxi Agricultural and Animal Husbandry Seed Farm, Shaanxi Fufeng, 722203, People's Republic of China
| | - Chuzhao Lei
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China
| | - Yu Jiang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China
| | - Eryao Wang
- Institute of Animal Husbandry, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, Henan, People's Republic of China.
| | - Yongzhen Huang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling Shaanxi, 712100, Shaanxi, People's Republic of China.
| |
Collapse
|
9
|
Maes M, Almulla AF, Zhou B, Algon AAA, Sodsai P. In major dysmood disorder, physiosomatic, chronic fatigue and fibromyalgia symptoms are driven by immune activation and increased immune-associated neurotoxicity. Sci Rep 2024; 14:7344. [PMID: 38538641 PMCID: PMC10973347 DOI: 10.1038/s41598-024-57350-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 11/12/2024] Open
Abstract
Major depressive disorder (MDD) is accompanied by activated neuro-immune pathways, increased physiosomatic and chronic fatigue-fibromyalgia (FF) symptoms. The most severe MDD phenotype, namely major dysmood disorder (MDMD), is associated with adverse childhood experiences (ACEs) and negative life events (NLEs) which induce cytokines/chemokines/growth factors. To delineate the impact of ACE + NLEs on physiosomatic and FF symptoms in first episode (FE)-MDMD, and examine whether these effects are mediated by immune profiles. ACEs, NLEs, physiosomatic and FF symptoms, and 48 cytokines/chemokines/growth factors were measured in 64 FE-MDMD patients and 32 normal controls. Physiosomatic, FF and gastro-intestinal symptoms belong to the same factor as depression, anxiety, melancholia, and insomnia. The first factor extracted from these seven domains is labeled the physio-affective phenome of depression. A part (59.0%) of the variance in physiosomatic symptoms is explained by the independent effects of interleukin (IL)-16 and IL-8 (positively), CCL3 and IL-1 receptor antagonist (inversely correlated). A part (46.5%) of the variance in physiosomatic (59.0%) symptoms is explained by the independent effects of interleukin (IL)-16, TNF-related apoptosis-inducing ligand (TRAIL) (positively) and combined activities of negative immunoregulatory cytokines (inversely associated). Partial least squares analysis shows that ACE + NLEs exert a substantial influence on the physio-affective phenome which are partly mediated by an immune network composed of interleukin-16, CCL27, TRAIL, macrophage-colony stimulating factor, and stem cell growth factor. The physiosomatic and FF symptoms of FE-MDMD are partly caused by immune-associated neurotoxicity due to T helper (Th)-1 polarization and M1 macrophage activation and relative lowered compensatory immunoregulatory protection.
Collapse
Affiliation(s)
- Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria.
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| | - Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China
| | - Ali Abbas Abo Algon
- Research Group of Organic Synthesis and Catalysis, University of Pannonia, Egyetem u. 10, Veszprém, 8200, Hungary
| | - Pimpayao Sodsai
- Department of Immunology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
10
|
Elmore A, Adhikari N, Hartley AE, Javier Aparicio H, Posner DC, Hemani G, Tilling K, Gaunt TR, Wilson P, Casas JP, Michael Gaziano J, Smith GD, Paternoster L, Cho K, Peloso GM. Protein identification for stroke progression via Mendelian Randomization in Million Veteran Program and UK Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.31.24302111. [PMID: 38352469 PMCID: PMC10863017 DOI: 10.1101/2024.01.31.24302111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Background Individuals who have experienced a stroke, or transient ischemic attack, face a heightened risk of future cardiovascular events. Identification of genetic and molecular risk factors for subsequent cardiovascular outcomes may identify effective therapeutic targets to improve prognosis after an incident stroke. Methods We performed genome-wide association studies (GWAS) for subsequent major adverse cardiovascular events (MACE) (Ncases=51,929, Ncntrl=39,980) and subsequent arterial ischemic stroke (AIS) Ncases=45,120, Ncntrl=46,789) after first incident stroke within the Million Veteran Program and UK Biobank. We then used genetic variants associated with proteins (pQTLs) to determine the effect of 1,463 plasma protein abundances on subsequent MACE using Mendelian randomization (MR). Results Two variants were significantly associated with subsequent cardiovascular events: rs76472767 (OR=0.75, 95% CI = 0.64-0.85, p= 3.69×10-08) with subsequent AIS and rs13294166 (OR=1.52, 95% CI = 1.37-1.67, p=3.77×10-08) with subsequent MACE. Using MR, we identified 2 proteins with an effect on subsequent MACE after a stroke: CCL27 (effect OR= 0.77, 95% CI = 0.66-0.88, adj. p=0.05), and TNFRSF14 (effect OR=1.42, 95% CI = 1.24-1.60, adj. p=0.006). These proteins are not associated with incident AIS and are implicated to have a role in inflammation. Conclusions We found evidence that two proteins with little effect on incident stroke appear to influence subsequent MACE after incident AIS. These associations suggest that inflammation is a contributing factor to subsequent MACE outcomes after incident AIS and highlights potential novel targets.
Collapse
Affiliation(s)
- Andrew Elmore
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Nimish Adhikari
- Veteran’s Affairs Healthcare System, Boston, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - April E Hartley
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Hugo Javier Aparicio
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Boston Medical Center, Boston, MA
| | | | - Gibran Hemani
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Kate Tilling
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Tom R Gaunt
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | | | - JP Casas
- Veteran’s Affairs Healthcare System, Boston, MA
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School
| | - John Michael Gaziano
- Veteran’s Affairs Healthcare System, Boston, MA
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School
| | - George Davey Smith
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Lavinia Paternoster
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol
| | - Kelly Cho
- Veteran’s Affairs Healthcare System, Boston, MA
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School
| | - Gina M Peloso
- Veteran’s Affairs Healthcare System, Boston, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| |
Collapse
|
11
|
Sigdel TK, Sur S, Boada P, McDermott SM, Arlehamn CSL, Murray KO, Bockenstedt LK, Kerwin M, Reed EF, Harris E, Stuart K, Peters B, Sesma A, Montgomery RR, Sarwal MM. Proteome Analysis for Inflammation Related to Acute and Convalescent Infection. Inflammation 2024; 47:346-362. [PMID: 37831367 PMCID: PMC10799112 DOI: 10.1007/s10753-023-01913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/30/2023] [Accepted: 09/24/2023] [Indexed: 10/14/2023]
Abstract
Infectious diseases are a significant burden in global healthcare. Pathogens engage with different host defense mechanisms. However, it is currently unknown if there are disease-specific immune signatures and/or if different pathogens elicit common immune-associated molecular entities to common therapeutic interventions. We studied patients enrolled through the Human Immunology Project Consortium (HIPC), which focuses on immune responses to various infections. Blood samples were collected and analyzed from patients during infection and follow-up time points at the convalescent stage. The study included samples from patients with Lyme disease (LD), tuberculosis (TB), malaria (MLA), dengue virus (DENV), and West Nile virus (WNV), as well as kidney transplant patients with cytomegalovirus (CMV) and polyomavirus (BKV) infections. Using an antibody-based assay, we quantified ~ 350 cell surface markers, cytokines, and chemokines involved in inflammation and immunity. Unique protein signatures were identified specific to the acute phase of infection irrespective of the pathogen type, with significant changes during convalescence. In addition, tumor necrosis factor receptor superfamily member 6 (TNR6), C-C Motif Chemokine Receptor 7 (CCR7), and C-C motif chemokine ligand-1 (CCL1) were increased in the acute and convalescent phases across all viral, bacterial, and protozoan compared to blood from healthy donors. Furthermore, despite the differences between pathogens, proteins were enriched in common biological pathways such as cell surface receptor signaling pathway and response to external stimulus. In conclusion, we demonstrated that irrespective of the pathogen type, there are common immunoregulatory and proinflammatory signals.
Collapse
Affiliation(s)
- Tara K Sigdel
- Division of Multi-Organ Transplantation, Department of Surgery, University of California San Francisco, 513 Parnassus Ave, Med Sciences Bldg, Room S1268, San Francisco, CA, 94143, USA
| | - Swastika Sur
- Division of Multi-Organ Transplantation, Department of Surgery, University of California San Francisco, 513 Parnassus Ave, Med Sciences Bldg, Room S1268, San Francisco, CA, 94143, USA
| | - Patrick Boada
- Division of Multi-Organ Transplantation, Department of Surgery, University of California San Francisco, 513 Parnassus Ave, Med Sciences Bldg, Room S1268, San Francisco, CA, 94143, USA
| | | | - Cecilia S Lindestam Arlehamn
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Maggie Kerwin
- Division of Multi-Organ Transplantation, Department of Surgery, University of California San Francisco, 513 Parnassus Ave, Med Sciences Bldg, Room S1268, San Francisco, CA, 94143, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eva Harris
- University of California Berkeley, Berkeley, CA, USA
| | - Ken Stuart
- Seattle Children Research Institute, Seattle, WA, USA
| | - Bjoern Peters
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ana Sesma
- Mount Sinai School of Medicine, New York, NY, USA
| | | | - Minnie M Sarwal
- Division of Multi-Organ Transplantation, Department of Surgery, University of California San Francisco, 513 Parnassus Ave, Med Sciences Bldg, Room S1268, San Francisco, CA, 94143, USA.
| |
Collapse
|
12
|
Lee MJ, Hammouda MB, Miao W, Okafor A, Jin Y, Sun H, Jain V, Markovtsov V, Diao Y, Gregory SG, Zhang JY. UBE2N is essential for maintenance of skin homeostasis and suppression of inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569631. [PMID: 38105982 PMCID: PMC10723344 DOI: 10.1101/2023.12.01.569631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
UBE2N, a Lys63-ubiquitin conjugating enzyme, plays critical roles in embryogenesis and immune system development and function. However, its roles in adult epithelial tissue homeostasis and pathogenesis are unclear. We generated conditional mouse models that deleted Ube2n in skin cells in a temporally and spatially controlled manner. We found that Ube2n-knockout (KO) in the adult skin keratinocytes induced a range of inflammatory skin defects characteristic of psoriatic and actinic keratosis. These included eczematous inflammation, epidermal and dermal thickening, parakeratosis, and increased immune cell infiltration, as well as signs of edema and blistering. Single cell transcriptomic analyses and RT-qPCR showed that Ube2n KO keratinocytes expressed elevated myeloid cell chemo-attractants such as Cxcl1 and Cxcl2 and decreased the homeostatic T lymphocyte chemo-attractant, Ccl27a. Consistently, the infiltrating immune cells of Ube2n-KO skin were predominantly myeloid-derived cells including neutrophils and M1-like macrophages that were highly inflammatory, as indicated by expression of Il1β and Il24. Pharmacological blockade of the IL-1 receptor associated kinases (IRAK1/4) alleviated eczema, epidermal and dermal thickening, and immune infiltration of the Ube2n mutant skin. Together, these findings highlight a key role of keratinocyte-UBE2N in maintenance of epidermal homeostasis and skin immunity and identify IRAK1/4 as potential therapeutic target for inflammatory skin disorders.
Collapse
Affiliation(s)
- Min Jin Lee
- Department of Dermatology, Duke University, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Wanying Miao
- Department of Dermatology, Duke University, Durham, NC, USA
| | - Arinze Okafor
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Yingai Jin
- Department of Dermatology, Duke University, Durham, NC, USA
| | - Huiying Sun
- Department of Dermatology, Duke University, Durham, NC, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Jennifer Y Zhang
- Department of Dermatology, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| |
Collapse
|
13
|
Zhang S, Wang Q, Ye J, Fan Q, Lin X, Gou Z, Azzam MM, Wang Y, Jiang S. Transcriptome and proteome profile of jejunum in chickens challenged with Salmonella Typhimurium revealed the effects of dietary bilberry anthocyanin on immune function. Front Microbiol 2023; 14:1266977. [PMID: 38053560 PMCID: PMC10694457 DOI: 10.3389/fmicb.2023.1266977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/13/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction The present study investigated the effects of bilberry anthocyanin (BA) on immune function when alleviating Salmonella Typhimurium (S. Typhimurium) infection in chickens. Methods A total of 180 newly hatched yellow-feathered male chicks were assigned to three groups (CON, SI, and SI + BA). Birds in CON and SI were fed a basal diet, and those in SI + BA were supplemented with 100 mg/kg BA for 18 days. Birds in SI and SI + BA received 0.5 ml suspension of S. Typhimurium (2 × 109 CFU/ml) by oral gavage at 14 and 16 days of age, and those in CON received equal volumes of sterile PBS. Results At day 18, (1) dietary BA alleviated weight loss of chickens caused by S. Typhimurium infection (P < 0.01). (2) Supplementation with BA reduced the relative weight of the bursa of Fabricius (P < 0.01) and jejunal villus height (P < 0.05) and increased the number of goblet cells (P < 0.01) and the expression of MUC2 (P < 0.05) in jejunal mucosa, compared with birds in SI. (3) Supplementation with BA decreased (P < 0.05) the concentration of immunoglobulins and cytokines in plasma (IgA, IL-1β, IL-8, and IFN-β) and jejunal mucosa (IgG, IgM, sIgA, IL-1β, IL-6, IL-8, TNF-α, IFN-β, and IFN-γ) of S. Typhimurium-infected chickens. (4) BA regulated a variety of biological processes, especially the defense response to bacteria and humoral immune response, and suppressed cytokine-cytokine receptor interaction and intestinal immune network for IgA production pathways by downregulating 6 immune-related proteins. Conclusion In summary, the impaired growth performance and disruption of jejunal morphology caused by S. Typhimurium were alleviated by dietary BA by affecting the expression of immune-related genes and proteins, and signaling pathways are related to immune response associated with immune cytokine receptors and production in jejunum.
Collapse
Affiliation(s)
- Sheng Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Qin Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Jinling Ye
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Qiuli Fan
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Xiajing Lin
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Zhongyong Gou
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Mahmoud M. Azzam
- Department of Animal Production College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Yibing Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Shouqun Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|