1
|
Datta D, Sulthana S, Strauss J, Puri A, Priyanka Bandi S, Singh S. Reconnoitring signaling pathways and exploiting innovative approaches tailoring multifaceted therapies for skin cancer. Int J Pharm 2024; 665:124719. [PMID: 39293575 DOI: 10.1016/j.ijpharm.2024.124719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Nowadays, skin cancer is widespread just like a varied malignant cancer which can cause serious health issues. Skin cancer, which encompasses malignant melanoma, basal cell carcinoma, and squamous cell carcinoma, is a prevalent form of cancer among humans. Due to its broad prevalence, financial burden, mortality rates, and cosmetic effects, it is a major public health issue. Skin cancer treatment involves surgery, chemotherapy, and radiation. Recently, personalized treatment in the fields of targeted therapies and precision medicine has been shown to diagnose early detection of every individual tumor by knowing their genetic and molecular characteristics. To target the molecular pathways responsible for tumor growth and reduce the damage to healthy tissue, new targeted therapies have emerged for melanoma, basal cell carcinoma, and squamous cell carcinoma. B-raf serine/threonine kinase (BRAF) and mitogen-activated protein kinase (MEK) inhibitors, immune checkpoint inhibitors, and precision medications have strong response rates to improve patient survival. Targeted therapeutics like nanocarriers have shown promising results by reducing skin irritation and protecting encapsulated therapeutics. These formulations have been shown to improve the transdermal permeability of anticancer drugs. The consideration of employing physical techniques to enhance the permeation of nanocarriers warrants attention to augment the dermal permeation of anticancer agents and facilitate targeted drug delivery within neoplastic cells. Targeted therapies face obstacles like resistance mechanisms and treatment strategy monitoring. Taken together, this review delves into the basic mechanisms of skin cancer, current treatment methods, drug resistance processes, and nano-based targeted techniques for cancer treatment. It will also delineate the challenges and perspectives in pre-clinical and clinical contexts.
Collapse
Affiliation(s)
- Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| | - Safiya Sulthana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Jordan Strauss
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614
| | - Sony Priyanka Bandi
- Loka Laboratories Private Limited, Technology Business Incubator, BITS Pilani Hyderabad Campus, Jawahar Nagar, Medchal 500078, Telangana, India.
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Yoon H, Koh D, Lim Y, Lee YH, Lee JK, Shin SY. Pyrazolines inhibiting the activity of the early growth response-1 DNA-binding domain. Bioorg Med Chem Lett 2024; 113:129952. [PMID: 39265893 DOI: 10.1016/j.bmcl.2024.129952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
To identify compounds inhibiting the activity of the Early Growth Response (EGR)-1 DNA-binding domain, thirty-seven pyrazolines were prepared and their EGR-1 DNA-binding activities were measured. Pharmacophores were derived based on quantitative structure-activity relationship calculations. As compound 2, 1-(5-(4-methoxyphenyl)-4,5-dihydro-1H-pyrazol-3-yl)naphthalen-2-ol, showed the best inhibitory effects against the activity of the EGR-1 DNA-binding domain, the binding mode between compound 2 and EGR-1 was elucidated using in silico docking. The pharmacophores were matched to the binding modes. Electrophoretic mobility shift assays confirmed that compound 2 dose-dependently inhibited TNFα-induced EGR-1-DNA complex formation in HaCaT cells. Reverse transcription-polymerase chain reaction demonstrated that compound 2 effectively reduced the mRNA expression of EGR-1-regulated inflammatory genes, including thymic stromal lymphopoietin (TSLP), interleukin (IL)-1β, IL-6, and IL-31, in TNFα-stimulated HaCaT cells. Therefore, compound 2 could be developed as an agent that inhibits the activity of the EGR-1 DNA-binding domain.
Collapse
Affiliation(s)
- Hyuk Yoon
- Division of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Young Han Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Jung Kul Lee
- Division of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
3
|
Sastri KT, Gupta NV, Kannan A, Dutta S, Ali M Osmani R, V B, Ramkishan A, S S. The next frontier in multiple sclerosis therapies: Current advances and evolving targets. Eur J Pharmacol 2024:177080. [PMID: 39491741 DOI: 10.1016/j.ejphar.2024.177080] [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: 05/28/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Recent advancements in research have significantly enhanced our comprehension of the intricate immune components that contribute to multiple sclerosis (MS) pathogenesis. By conducting an in-depth analysis of complex molecular interactions involved in the immunological cascade of the disease, researchers have successfully identified novel therapeutic targets, leading to the development of innovative therapies. Leveraging pioneering technologies in proteomics, genomics, and the assessment of environmental factors has expedited our understanding of the vulnerability and impact of these factors on the progression of MS. Furthermore, these advances have facilitated the detection of significant biomarkers for evaluating disease activity. By integrating these findings, researchers can design novel molecules to identify new targets, paving the way for improved treatments and enhanced patient care. Our review presents recent discoveries regarding the pathogenesis of MS, highlights their genetic implications, and proposes an insightful approach for engaging with newer therapeutic targets in effectively managing this debilitating condition.
Collapse
Affiliation(s)
- K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - Anbarasu Kannan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - Balamuralidhara V
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - A Ramkishan
- Deputy Drugs Controller (India), Central Drugs Standard Control Organization, Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | | |
Collapse
|
4
|
Schloesser L, Klose SM, Mauschitz MM, Abdullah Z, Finger RP. The role of immune modulators in age-related macular degeneration. Surv Ophthalmol 2024; 69:851-869. [PMID: 39097172 DOI: 10.1016/j.survophthal.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We provide an overview of the expanding literature on the role of cytokines and immune mediators in pathophysiology of age-related macular degeneration (AMD). Although many immunological mediators have been linked to AMD pathophysiology, the broader mechanistic picture remains unclear with substantial variations in the levels of evidence supporting these mediators. Therefore, we reviewed the literature considering the varying levels of supporting evidence. A Medical Subject Headings (MeSH) term-based literature research was conducted in September, 2023, consisting of the MeSH terms "cytokine" and "Age-related macular degeneration" connected by the operator "AND". After screening the publications by title, abstract, and full text, a total of 146 publications were included. The proinflammatory cytokines IL-1β (especially in basic research studies), IL-6, IL-8, IL-18, TNF-α, and MCP-1 are the most extensively characterised cytokines/chemokines, highlighting the role of local inflammasome activation and altered macrophage function in the AMD pathophysiology. Among the antiinflammatory mediators IL-4, IL-10, and TGF-β were found to be the most extensively characterised, with IL-4 driving and IL-10 and TGF-β suppressing disease progression. Despite the extensive literature on this topic, a profound understanding of AMD pathophysiology has not yet been achieved. Therefore, further studies are needed to identify potential therapeutic targets, followed by clinical studies.
Collapse
Affiliation(s)
- Lukas Schloesser
- Department of Ophthalmology, University of Bonn, Bonn, Germany; Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Sara M Klose
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Asia-Pacific Centre for Animal Health, Faculty of Science, University of Melbourne, Melbourne, Australia
| | | | - Zeinab Abdullah
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Robert P Finger
- Department of Ophthalmology, University of Bonn, Bonn, Germany; Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
5
|
Charlès LM, von Reiterdank IF, Lancia HH, Shamlou AA, Berkane Y, Rosales I, Mink van der Molen AB, Coert J, Cetrulo CL, Lellouch AG, Uygun K. Effect of Subnormothermic Machine Perfusion on the Preservation of Vascularized Composite Allografts After Prolonged Warm Ischemia. Transplantation 2024; 108:2222-2232. [PMID: 38722685 PMCID: PMC11518650 DOI: 10.1097/tp.0000000000005035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Warm ischemia time (WIT) and ischemia-reperfusion injury are limiting factors for vascularized composite allograft (VCA) transplantation. Subnormothermic machine perfusion (SNMP) has demonstrated the potential to extend WIT in organ transplantation. This study evaluates the effect of SNMP on VCA viability after prolonged WIT. METHODS Rat hindlimbs underwent WIT for 30, 45, 60, 120, 150, or 210 min, followed by 3-h SNMP. Monitoring of perfusion parameters and outflow determined the maximum WIT compatible with limb viability after SNMP. Thereafter, 2 groups were assessed: a control group with inbred transplantation (Txp) after 120 min of WIT and an experimental group that underwent WIT + SNMP + Txp. Graft appearance, blood gas, cytokine levels, and histology were assessed for 21 d. RESULTS Based on potassium levels, the limit of WIT compatible with limb viability after SNMP is 120 min. Before this limit, SNMP reduces potassium and lactate levels of WIT grafts to the same level as fresh grafts. In vivo, the control group presented 80% graft necrosis, whereas the experimental group showed no necrosis, had better healing ( P = 0.0004), and reduced histological muscle injury ( P = 0.012). Results of blood analysis revealed lower lactate, potassium levels, and calcium levels ( P = 0.048) in the experimental group. Both groups presented an increase in interleukin (IL)-10 and IL-1b/IL-1F2 with a return to baseline after 7 to 14 d. CONCLUSIONS Our study establishes the limit of WIT compatible with VCA viability and demonstrates the effectiveness of SNMP in restoring a graft after WIT ex vivo and in vivo, locally and systemically.
Collapse
Affiliation(s)
- Laura M. Charlès
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
| | - Irina Filz von Reiterdank
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hyshem H Lancia
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
| | - Austin Alana Shamlou
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
| | - Yanis Berkane
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
- Department of Plastic, Reconstructive and Aesthetic Surgery, Rennes University Hospital Center (CHU de Rennes), Rennes University, Rennes, France
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Aebele B. Mink van der Molen
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J.H Coert
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Curtis L Cetrulo
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
| | - Alexandre G Lellouch
- Vascularized Composite Allotransplantation Laboratory, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
| | - Korkut Uygun
- Harvard Medical School, Boston, MA
- Shriners Children’s Boston, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
6
|
Beucher L, Gabillard-Lefort C, Baris OR, Mialet-Perez J. Monoamine oxidases: A missing link between mitochondria and inflammation in chronic diseases ? Redox Biol 2024; 77:103393. [PMID: 39405979 PMCID: PMC11525629 DOI: 10.1016/j.redox.2024.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
The role of mitochondria spans from the regulation of the oxidative phosphorylation, cell metabolism and survival/death pathways to a more recently identified function in chronic inflammation. In stress situations, mitochondria release some pro-inflammatory mediators such as ATP, cardiolipin, reactive oxygen species (ROS) or mitochondrial DNA, that are believed to participate in chronic diseases and aging. These mitochondrial Damage-Associated Molecular Patterns (mito-DAMPs) can modulate specific receptors among which TLR9, NLRP3 and cGAS-STING, triggering immune cells activation and sterile inflammation. In order to counter the development of chronic diseases, a better understanding of the underlying mechanisms of low grade inflammation induced by mito-DAMPs is needed. In this context, monoamine oxidases (MAO), the mitochondrial enzymes that degrade catecholamines and serotonin, have recently emerged as potent regulators of chronic inflammation in obesity-related disorders, cardiac diseases, cancer, rheumatoid arthritis and pulmonary diseases. The role of these enzymes in inflammation embraces their action in both immune and non-immune cells, where they regulate monoamines levels and generate toxic ROS and aldehydes, as by-products of enzymatic reaction. Here, we discuss the more recent advances on the role and mechanisms of action of MAOs in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Lise Beucher
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France
| | | | - Olivier R Baris
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France
| | - Jeanne Mialet-Perez
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France.
| |
Collapse
|
7
|
Gyorffy VJ, Dwivedi DJ, Liaw PC, Fox-Robichaud AE, Tsang JLY, Binnie A. Impact of sample processing delays on plasma markers of inflammation, chemotaxis, cell death, and blood coagulation. PLoS One 2024; 19:e0311921. [PMID: 39480839 PMCID: PMC11527306 DOI: 10.1371/journal.pone.0311921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Biosampling studies in critically ill patients traditionally involve bedside collection of samples followed by local processing (ie. centrifugation, aliquotting, and freezing) and storage. However, community hospitals, which care for the majority of Canadian patients, often lack the infrastructure for local processing and storage of specimens. A potential solution is a "simplified" biosampling protocol whereby blood samples are collected at the bedside and then shipped to a central site for processing and storage. One potential limitation of this approach is that delayed processing may alter sample characteristics. OBJECTIVE To determine whether delays in blood sample processing affect the stability of cytokines (IL-6, TNF, IL-10, IFN-γ), chemokines (IL-8, IP-10, MCP-1, MCP-4, MIP-1α, MIP-1β), cell-free DNA (cfDNA) (released by dying cells), and blood clotting potential in human blood samples. METHODS Venous blood was collected into EDTA and citrate sample tubes and stored at room temperature (RT) or 4°C for progressive intervals up to 72 hours, prior to processing. Plasma cytokines and chemokines were quantified using single or multiplex immunoassays. cfDNA was measured using Picogreen DNA Quantification. Blood clotting potential was measured using a thrombin generation assay. RESULTS Blood samples were collected from 9 intensive care unit (ICU) patients and 7 healthy volunteers. Admission diagnoses for the ICU patients included sepsis, trauma, ruptured abdominal aortic aneurysm, intracranial hemorrhage, gastrointestinal bleed, and hyperkalemia. After pre-processing delays of up to 72 hours at RT or 4°C, no significant changes were observed in plasma cytokines, chemokines, cfDNA, or thrombin formation. CONCLUSIONS Delayed sample processing for up to 72 hours at either RT or 4°C did not significantly affect cytokines, chemokines, cfDNA, or blood clotting potential in plasma samples from healthy volunteers and ICU patients. A "simplified" biosampling protocol is a feasible solution for conducting biosampling research at hospitals without local processing capacity.
Collapse
Affiliation(s)
- Vanessa J. Gyorffy
- Faculty of Arts and Science, McMaster University, Hamilton, ON, Canada
- Niagara Health Knowledge Institute, St. Catharines, ON, Canada
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
| | - Dhruva J. Dwivedi
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Patricia C. Liaw
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alison E. Fox-Robichaud
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer L. Y. Tsang
- Niagara Health Knowledge Institute, St. Catharines, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexandra Binnie
- Critical Care Department, William Osler Health System, Etobicoke, ON, Canada
| |
Collapse
|
8
|
Kersh AE, Sati S, Huang J, Murphy C, Ahart O, Leung TH. CXCL9, CXCL10, and CCL19 synergistically recruit T lymphocytes to skin in lichen planus. JCI Insight 2024; 9:e179899. [PMID: 39190494 DOI: 10.1172/jci.insight.179899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Lichen planus (LP) is a chronic, debilitating, inflammatory disease of the skin and mucous membranes that affects 1%-2% of Americans. Its molecular pathogenesis remains poorly understood, and there are no FDA-approved treatments. We performed single-cell RNA sequencing on paired blood and skin samples (lesional and nonlesional tissue) from 7 patients with LP. We discovered that LP keratinocytes and fibroblasts specifically secrete a combination of CXCL9, CXCL10, and CCL19 cytokines. Using an in vitro migration assay with primary human T cells, we demonstrated that CCL19 in combination with either of the other 2 cytokines synergistically enhanced recruitment of CD8+ T cells more than any individual cytokine. Moreover, exhausted T cells in lesional LP skin secreted CXCL13, which, along with CCL19, also enhanced recruitment of T cells, suggesting a feed-forward loop in LP. Finally, LP blood revealed decreased circulating naive CD8+ T cells compared with that in healthy volunteers, consistent with recruitment to skin. Molecular analysis of LP skin and blood samples increased our understanding of disease pathogenesis and identified CCL19 as a new therapeutic target for treatment.
Collapse
Affiliation(s)
- Anna E Kersh
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Satish Sati
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jianhe Huang
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christina Murphy
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Olivia Ahart
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas H Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Gao D, Zou B, Zhu K, Bi S, Zhang W, Yang X, Lai J, Liang G, Pan P. Enhancing Th17 cells drainage through meningeal lymphatic vessels alleviate neuroinflammation after subarachnoid hemorrhage. J Neuroinflammation 2024; 21:269. [PMID: 39428510 PMCID: PMC11492769 DOI: 10.1186/s12974-024-03252-y] [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: 08/23/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disorder primarily caused by the rupture of aneurysm, which results in a high mortality rate and consequently imposes a significant burden on society. The occurrence of SAH initiates an immune response that further exacerbates brain damage. The acute inflammatory reaction subsequent to SAH plays a crucial role in determining the prognosis. Th17 cells, a subset of T cells, are related to the brain injury following SAH, and it is unclear how Th17 cells are cleared in the brain. Meningeal lymphatic vessels are a newly discovered intracranial fluid transport system that has been shown to drain large molecules and immune cells to deep cervical lymph nodes. There is limited understanding of the role of the meningeal lymphatic system in SAH. The objective of this research is to explore the impact and underlying mechanism of drainage Th17 cells by meningeal lymphatics on SAH. METHODS Treatments to manipulate meningeal lymphatic function and the CCR7-CCL21 pathway were administered, including laser ablation, injection of VEGF-C geneknockout, and protein injection. Mouse behavior was assessed using the balance beam experiment and the modified Garcia scoring system. Flow cytometry, enzyme-linked immunosorbent assays (ELISA), and immunofluorescence staining were used to study the impact of meningeal lymphatic on SAH drainage. Select patients with unruptured and ruptured aneurysms in our hospital as the control group and the SAH group, with 7 cases in each group. Peripheral blood and cerebrospinal fluid (CSF) samples were assessed by ELISA and flow cytometry. RESULTS Mice with SAH showed substantial behavioral abnormalities and brain damage in which immune cells accumulated in the brain. Laser ablation of the meningeal lymphatic system or knockout of the CCR7 gene leads to Th17 cell aggregation in the meninges, resulting in a decreased neurological function score and increased levels of inflammatory factors. Injection of VEGF-C or CCL21 protein promotes Th17 cell drainage to lymph nodes, an increased neurological function score, and decreased levels of inflammatory factors. Clinical blood and CSF results showed that inflammatory factors in SAH group were significantly increased. The number of Th17 cells in the SAH group was significantly higher than the control group. Clinical results confirmed Th17 cells aggravated the level of neuroinflammation after SAH. CONCLUSION This study shows that improving the drainage of Th17 cells by meningeal lymphatics via the CCR7-CCL21 pathway can reduce brain damage and improve behavior in the SAH mouse model. This could lead to new treatment options for SAH.
Collapse
Affiliation(s)
- Dandan Gao
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
- China Medical University, Shenyang, Liaoning, China
| | - Bin Zou
- Department of Aanesthesiology, General Hospital of Northern Theater Command, Shenyang, China
- Dalian Medical University, Dalian, China
| | - Kunyuan Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Shijun Bi
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Wenxu Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xinyu Yang
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jieyu Lai
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Pengyu Pan
- Department of Neurosurgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
10
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
11
|
Vieira SF, Gonçalves SM, Gonçalves VMF, Tiritan ME, Cunha C, Carvalho A, Reis RL, Ferreira H, Neves NM. Evaluation of Echinacea purpurea Extracts as Immunostimulants: Impact on Macrophage Activation. PLANTA MEDICA 2024. [PMID: 39419081 DOI: 10.1055/a-2436-9664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Echinacea purpurea has been traditionally used to strengthen the immune system. Therefore, herein, we investigated the potential of E. purpurea aqueous extracts (AEs) obtained from flowers (F), leaves (L), or roots (R) as an immune booster in human primary monocyte-derived macrophages (hMDMs). Additionally, to identify the main class of compounds (phenolic/carboxylic acids vs. alkylamides) responsible for the bioactivity, the three AEs were fractioned by semi-preparative high-performance liquid chromatography (HPLC). The AEs and the isolated phenolic/carboxylic acidic fractions were not cytotoxic for hMDMs for all tested concentrations, as confirmed by the metabolic activity and DNA content assays. Moreover, AE drastically induced the production of the interleukin (IL)-6 and tumor necrosis factor (TNF)-α, with a minimal effect on IL-1β and prostaglandin E2 (PGE2), supporting their potential for macrophage activation. Interestingly, in the presence of the phenolic/carboxylic acidic fractions, this efficacy considerably decreased, suggesting a complementary effect between compounds. AE also triggered the phosphorylation of the extracellular signal-regulated kinase (ERK) 1/2 and p38 signaling pathways and upregulated the cyclooxygenase (COX)-2 expression in hMDMs. Overall, AE-F was demonstrated to be the most powerful immunostimulant extract that can be related to their higher number in identified bioactive compounds compared to AE-L and AE-R. These results highlight the efficiency of E. purpurea AE to enhance the function of a key cell type of the immune system and their potential as immunostimulant formulations for patients with a compromised immune system due to certain diseases (e.g., acquired immunodeficiencies) and treatments (e.g., chemotherapy).
Collapse
Affiliation(s)
- Sara F Vieira
- 3B's Research Group, I3BS - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Samuel M Gonçalves
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Virgínia M F Gonçalves
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, Gandra, Portugal
- UNIPRO - Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), CESPU, Gandra, Portugal
| | - Maria E Tiritan
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, Gandra, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Matosinhos, Portugal
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | - Cristina Cunha
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Agostinho Carvalho
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3BS - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helena Ferreira
- 3B's Research Group, I3BS - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno M Neves
- 3B's Research Group, I3BS - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
12
|
Yau A, Sands I, Zhang W, Chen Y. Injectable Janus Base Nanomatrix (JBNm) in Maintaining Long-Term Homeostasis of Regenerated Cartilage for Tissue Chip Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.05.616785. [PMID: 39416084 PMCID: PMC11482866 DOI: 10.1101/2024.10.05.616785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Engineered cartilage tissues have wide applications in in vivo cartilage repair as well as in vitro models, such as cartilage-on-a-chip or cartilage tissue chips. Currently, most cartilage tissue engineering approaches focus on promoting chondrogenesis of stem cells to produce regenerated cartilage. However, this regenerated cartilage can dedifferentiate into fibrotic tissue or further differentiate into hypertrophic or calcified cartilage. One of the most challenging objectives in cartilage tissue engineering is to maintain long-term cartilage homeostasis. Since the microenvironment of engineered cartilage tissue is crucial for stem cell adhesion, proliferation, differentiation, and function, we aim to develop a novel scaffold that can maintain the long-term homeostasis of regenerated cartilage. Therefore, we developed a library of Janus base nanomatrices (JBNms), composed of DNA-inspired Janus nanotubes (JBNts) as well as cartilage extracellular matrix (ECM) proteins. The JBNms were developed to selectively promote chondro-lineage cell functions while inhibiting bone and endothelial cell growth. More importantly, the JBNm can effectively promote chondrogenesis while inhibiting hypertrophy, osteogenesis, angiogenesis, and dedifferentiation. Additionally, the JBNm is injectable, forming a solid scaffold suitable for producing and maintaining regenerated cartilage tissue in microfluidic chips, making it ideal for tissue chip applications. In this study, we successfully created cartilage tissue chips using JBNms. These chips can model cartilage tissue even after long-term culture and can also mimic arthritis progression, making them useful for drug screening. Thus, we have developed a novel nanomaterial approach for improved cartilage tissue engineering and cartilage tissue chip applications.
Collapse
Affiliation(s)
| | | | - Wuxia Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
13
|
Padovani-Claudio DA, Morales MS, Smith TE, Ontko CD, Namburu NS, Palmer SA, Jhala MG, Ramos CJ, Capozzi ME, McCollum GW, Penn JS. Induction, amplification, and propagation of diabetic retinopathy-associated inflammatory cytokines between human retinal microvascular endothelial and Müller cells and in the mouse retina. Cell Signal 2024; 124:111454. [PMID: 39384004 DOI: 10.1016/j.cellsig.2024.111454] [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: 07/30/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Ocular levels of IL-1β, TNFα, IL-8, and IL-6 correlate with progression of diabetic retinopathy (DR). Müller cells (MC), which are crucial to maintaining retinal homeostasis, are targets and sources of these cytokines. We explored the relative capacities of these four DR-associated cytokines to amplify inflammatory signal expression both in and between human MC (hMC) and retinal microvascular endothelial cells (hRMEC) and in the mouse retina. Of the four cytokines, IL-1β was the most potent stimulus of transcriptomic alterations in hMC and hRMEC in vitro, as well as in the mouse retina after intravitreal injection in vivo. Stimulation with IL-1β significantly induced expression of all four transcripts in hMC and hRMEC. TNFα significantly induced expression of some, but not all, of the four transcripts in each cell, while neither IL-8 nor IL-6 showed significant induction in either cell. Similarly, conditioned media (CM) derived from hMC or hRMEC treated with IL-1β, but not TNFα, upregulated inflammatory cytokine transcripts in the reciprocal cell type. hRMEC responses to hMC-derived CM were dependent on IL-1R activation. In addition, we observed a correlation between cytokine expression changes following direct and CM stimulation and NFκB-p65 nuclear translocation in both hMC and hRMEC. Finally, in mice, intravitreal injections of IL-1β, but not TNFα, induced retinal expression of Il1b and CXCL8 homologues Cxcl1, Cxcl2, Cxcl3, and Cxcl5, encoding pro-angiogenic chemokines. Our results suggest that expression of IL-1β, TNFα, IL-8, and IL-6 may be initiated, propagated, and sustained by autocrine and paracrine signals in hRMEC and hMC through a process involving IL-1β and NFκB. Targeting these signals may help thwart inflammatory amplification, preventing progression to vision-threatening stages and preserving sight.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Monica S Morales
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Taylor E Smith
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Cayla D Ontko
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 1161 21st Ave S., Nashville, TN 37232, USA.
| | - Neeraj S Namburu
- College of Arts and Sciences, Vanderbilt University, 2400 Vanderbilt Pl., Nashville, TN 37232, USA.
| | - Samuel A Palmer
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Marvarakumari G Jhala
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - Megan E Capozzi
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| | - Gary W McCollum
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 1161 21st Ave S., Nashville, TN 37232, USA.
| |
Collapse
|
14
|
Huang H, Cao D, Hu Y, He Q, Zhao X, Chen L, Lin S, Luo X, Ye Y, Liao J, Zou H, Zou D. Exploring Infantile Epileptic Spasm Syndrome: A Proteomic Analysis of Plasma Using the Data-Independent Acquisition Approach. J Proteome Res 2024; 23:4316-4326. [PMID: 38857073 PMCID: PMC11459594 DOI: 10.1021/acs.jproteome.4c00298] [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: 04/11/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024]
Abstract
This study aimed to identify characteristic proteins in infantile epileptic spasm syndrome (IESS) patients' plasma, offering insights into potential early diagnostic biomarkers and its underlying causes. Plasma samples were gathered from 60 patients with IESS and 40 healthy controls. Data-independent acquisition proteomic analysis was utilized to identify differentially expressed proteins (DEPs). These DEPs underwent functional annotation through Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Gene set enrichment analysis (GSEA) was employed for both GO (GSEA-GO) and KEGG (GSEA-KEGG) analyses to examine the gene expression profiles. Receiver operating characteristic (ROC) curves assessed biomarkers' discriminatory capacity. A total of 124 DEPs were identified in IESS patients' plasma, mainly linked to pathways, encompassing chemokines, cytokines, and oxidative detoxification. GSEA-GO and GSEA-KEGG analyses indicated significant enrichment of genes associated with cell migration, focal adhesion, and phagosome pathways. ROC curve analysis demonstrated that the combination of PRSS1 and ACTB, PRSS3, ACTB, and PRSS1 alone exhibited AUC values exceeding 0.7. This study elucidated the significant contribution of cytokines, chemokines, oxidative detoxification, and phagosomes to the IESS pathogenesis. The combination of PRSS1 and ACTB holds promise as biomarkers for the early diagnosis of IESS.
Collapse
Affiliation(s)
- Haohua Huang
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
- Shenzhen
Pediatrics Institute of Shantou University Medical College, Shenzhen 518000 Guangdong, China
| | - Dezhi Cao
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Yan Hu
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Qianqian He
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Xia Zhao
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Li Chen
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Sufang Lin
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Xufeng Luo
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Yuanzhen Ye
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Jianxiang Liao
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Huafang Zou
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| | - Dongfang Zou
- Epilepsy
Center and Department of Neurology, Shenzhen
Children’s Hospital, Shenzhen 518000 Guangdong, China
| |
Collapse
|
15
|
Alves PT, de Souza AG, Bastos VAF, Miguel EL, Ramos ACS, Cameron LC, Goulart LR, Cunha TM. The Modulation of Septic Shock: A Proteomic Approach. Int J Mol Sci 2024; 25:10641. [PMID: 39408970 PMCID: PMC11476436 DOI: 10.3390/ijms251910641] [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: 08/28/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Sepsis poses a significant challenge due its lethality, involving multiple organ dysfunction and impaired immune responses. Among several factors affecting sepsis, monocytes play a crucial role; however, their phenotype, proteomic profile, and function in septic shock remain unclear. Our aim was to fully characterize the subpopulations and proteomic profiles of monocytes seen in septic shock cases and discuss their possible impact on the disease. Peripheral blood monocyte subpopulations were phenotype based on CD14/CD16 expression by flow cytometry, and proteins were extracted from the monocytes of individuals with septic shock and healthy controls to identify changes in the global protein expression in these cells. Analysis using 2D-nanoUPLC-UDMSE identified 67 differentially expressed proteins in shock patients compared to controls, in which 44 were upregulated and 23 downregulated. These proteins are involved in monocyte reprogramming, immune dysfunction, severe hypotension, hypo-responsiveness to vasoconstrictors, vasodilation, endothelial dysfunction, vascular injury, and blood clotting, elucidating the disease severity and therapeutic challenges of septic shock. This study identified critical biological targets in monocytes that could serve as potential biomarkers for the diagnosis, prognosis, and treatment of septic shock, providing new insights into the pathophysiology of the disease.
Collapse
Affiliation(s)
- Patrícia Terra Alves
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Aline Gomes de Souza
- Department of Medical Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14040-900, SP, Brazil;
| | - Victor Alexandre F. Bastos
- Laboratory of Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Eduarda L. Miguel
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - Augusto César S. Ramos
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - L. C. Cameron
- Arthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada;
- Lorraine Protein Biochemistry Group, Graduate Program in Neurology, Gaffrée e Guinle University Hospital, Rio de Janeiro 20270-004, RJ, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Thúlio M. Cunha
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| |
Collapse
|
16
|
Yamaguchi H, Nishimura Y, Matsuse D, Sekiya H, Masaki K, Tanaka T, Saiga T, Harada M, Kira YI, Dickson DW, Fujishima K, Matsuo E, Tanaka KF, Yamasaki R, Isobe N, Kira JI. A rapidly progressive multiple system atrophy-cerebellar variant model presenting marked glial reactions with inflammation and spreading of α-synuclein oligomers and phosphorylated α-synuclein aggregates. Brain Behav Immun 2024; 121:122-141. [PMID: 38986725 DOI: 10.1016/j.bbi.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/30/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple system atrophy (MSA) is a severe α-synucleinopathy facilitated by glial reactions; the cerebellar variant (MSA-C) preferentially involves olivopontocerebellar fibres with conspicuous demyelination. A lack of aggressive models that preferentially involve olivopontocerebellar tracts in adulthood has hindered our understanding of the mechanisms of demyelination and neuroaxonal loss, and thus the development of effective treatments for MSA. We therefore aimed to develop a rapidly progressive mouse model that recaptures MSA-C pathology. We crossed Plp1-tTA and tetO-SNCA*A53T mice to generate Plp1-tTA::tetO-SNCA*A53T bi-transgenic mice, in which human A53T α-synuclein-a mutant protein with enhanced aggregability-was specifically produced in the oligodendrocytes of adult mice using Tet-Off regulation. These bi-transgenic mice expressed mutant α-synuclein from 8 weeks of age, when doxycycline was removed from the diet. All bi-transgenic mice presented rapidly progressive motor deterioration, with wide-based ataxic gait around 22 weeks of age and death around 30 weeks of age. They also had prominent demyelination in the brainstem/cerebellum. Double immunostaining demonstrated that myelin basic protein was markedly decreased in areas in which SM132, an axonal marker, was relatively preserved. Demyelinating lesions exhibited marked ionised calcium-binding adaptor molecule 1-, arginase-1-, and toll-like receptor 2-positive microglial reactivity and glial fibrillary acidic protein-positive astrocytic reactivity. Microarray analysis revealed a strong inflammatory response and cytokine/chemokine production in bi-transgenic mice. Neuronal nuclei-positive neuronal loss and patchy microtubule-associated protein 2-positive dendritic loss became prominent at 30 weeks of age. However, a perceived decrease in tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta in bi-transgenic mice compared with wild-type mice was not significant, even at 30 weeks of age. Wild-type, Plp1-tTA, and tetO-SNCA*A53T mice developed neither motor deficits nor demyelination. In bi-transgenic mice, double immunostaining revealed human α-synuclein accumulation in neurite outgrowth inhibitor A (Nogo-A)-positive oligodendrocytes beginning at 9 weeks of age; its expression was further increased at 10 to 12 weeks, and these increased levels were maintained at 12, 24, and 30 weeks. In an α-synuclein-proximity ligation assay, α-synuclein oligomers first appeared in brainstem oligodendrocytes as early as 9 weeks of age; they then spread to astrocytes, neuropil, and neurons at 12 and 16 weeks of age. α-Synuclein oligomers in the brainstem neuropil were most abundant at 16 weeks of age and decreased thereafter; however, those in Purkinje cells successively increased until 30 weeks of age. Double immunostaining revealed the presence of phosphorylated α-synuclein in Nogo-A-positive oligodendrocytes in the brainstem/cerebellum as early as 9 weeks of age. In quantitative assessments, phosphorylated α-synuclein gradually and successively accumulated at 12, 24, and 30 weeks in bi-transgenic mice. By contrast, no phosphorylated α-synuclein was detected in wild-type, tetO-SNCA*A53T, or Plp1-tTA mice at any age examined. Pronounced demyelination and tubulin polymerisation, promoting protein-positive oligodendrocytic loss, was closely associated with phosphorylated α-synuclein aggregates at 24 and 30 weeks of age. Early inhibition of mutant α-synuclein expression by doxycycline diet at 23 weeks led to fully recovered demyelination; inhibition at 27 weeks led to persistent demyelination with glial reactions, despite resolving phosphorylated α-synuclein aggregates. In conclusion, our bi-transgenic mice exhibited progressively increasing demyelination and neuroaxonal loss in the brainstem/cerebellum, with rapidly progressive motor deterioration in adulthood. These mice showed marked microglial and astrocytic reactions with inflammation that was closely associated with phosphorylated α-synuclein aggregates. These features closely mimic human MSA-C pathology. Notably, our model is the first to suggest that α-synuclein oligomers may spread from oligodendrocytes to neurons in transgenic mice with human α-synuclein expression in oligodendrocytes. This model of MSA is therefore particularly useful for elucidating the in vivo mechanisms of α-synuclein spreading from glia to neurons, and for developing therapies that target glial reactions and/or α-synuclein oligomer spreading and aggregate formation in MSA.
Collapse
Affiliation(s)
- Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka, Japan.
| | - Yuji Nishimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Tatsunori Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Sumitomo Pharma Co., Ltd., Osaka, Japan.
| | - Toru Saiga
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaya Harada
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yuu-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | - Kei Fujishima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Eriko Matsuo
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Translational Neuroscience Research Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan; Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, Fukuoka, Japan.
| |
Collapse
|
17
|
Flinkenflügel K, Gruber M, Meinert S, Thiel K, Winter A, Goltermann J, Usemann P, Brosch K, Stein F, Thomas-Odenthal F, Wroblewski A, Pfarr JK, David FS, Beins EC, Grotegerd D, Hahn T, Leehr EJ, Dohm K, Bauer J, Forstner AJ, Nöthen MM, Jamalabadi H, Straube B, Alexander N, Jansen A, Witt SH, Rietschel M, Nenadić I, van den Heuvel MP, Kircher T, Repple J, Dannlowski U. The interplay between polygenic score for tumor necrosis factor-α, brain structural connectivity, and processing speed in major depression. Mol Psychiatry 2024; 29:3151-3159. [PMID: 38693319 PMCID: PMC11449800 DOI: 10.1038/s41380-024-02577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Reduced processing speed is a core deficit in major depressive disorder (MDD) and has been linked to altered structural brain network connectivity. Ample evidence highlights the involvement of genetic-immunological processes in MDD and specific depressive symptoms. Here, we extended these findings by examining associations between polygenic scores for tumor necrosis factor-α blood levels (TNF-α PGS), structural brain connectivity, and processing speed in a large sample of MDD patients. Processing speed performance of n = 284 acutely depressed, n = 177 partially and n = 198 fully remitted patients, and n = 743 healthy controls (HC) was estimated based on five neuropsychological tests. Network-based statistic was used to identify a brain network associated with processing speed. We employed general linear models to examine the association between TNF-α PGS and processing speed. We investigated whether network connectivity mediates the association between TNF-α PGS and processing speed. We identified a structural network positively associated with processing speed in the whole sample. We observed a significant negative association between TNF-α PGS and processing speed in acutely depressed patients, whereas no association was found in remitted patients and HC. The mediation analysis revealed that brain connectivity partially mediated the association between TNF-α PGS and processing speed in acute MDD. The present study provides evidence that TNF-α PGS is associated with decreased processing speed exclusively in patients with acute depression. This association was partially mediated by structural brain connectivity. Using multimodal data, the current findings advance our understanding of cognitive dysfunction in MDD and highlight the involvement of genetic-immunological processes in its pathomechanisms.
Collapse
Grants
- WI 3439/3-1, WI 3439/3-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- RI 908/11-1, RI 908/11-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- JA 1890/7-1, JA 1890/7-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- EP-C-16-015 EPA
- DA1151/5-1, DA1151/5-2, DA1151/11‑1 DA1151/6-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- NO 246/10-1, NO 246/10-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- HA7070/2-2, HA7070/3, HA7070/4 Deutsche Forschungsgemeinschaft (German Research Foundation)
- STR 1146/18-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- ERC-COG 101001062, VIDI-452-16-015 Nederlandse Organisatie voor Wetenschappelijk Onderzoek (Netherlands Organisation for Scientific Research)
- KI 588/14-1, KI 588/14-2, KI 588/22-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- Interdisziplinäres Zentrum für Klinische Forschung, medizinische Fakultät, Münster (Dan3/012/17)
- Innovative medizinische Forschung Münster (IMF): RE111604, RE111722, RE 221707
Collapse
Affiliation(s)
- Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Marius Gruber
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Adrian Wroblewski
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Julia-Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Eva C Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jochen Bauer
- Department of Radiology, University of Münster, Münster, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
- Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Martijn P van den Heuvel
- Connectome Lab, Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Child Psychiatry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.
| |
Collapse
|
18
|
Zhang C, Qiu M, Fu H. Oligodendrocytes in central nervous system diseases: the effect of cytokine regulation. Neural Regen Res 2024; 19:2132-2143. [PMID: 38488548 PMCID: PMC11034588 DOI: 10.4103/1673-5374.392854] [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: 07/31/2023] [Revised: 11/08/2023] [Accepted: 12/06/2023] [Indexed: 04/24/2024] Open
Abstract
Cytokines including tumor necrosis factor, interleukins, interferons, and chemokines are abundantly produced in various diseases. As pleiotropic factors, cytokines are involved in nearly every aspect of cellular functions such as migration, survival, proliferation, and differentiation. Oligodendrocytes are the myelin-forming cells in the central nervous system and play critical roles in the conduction of action potentials, supply of metabolic components for axons, and other functions. Emerging evidence suggests that both oligodendrocytes and oligodendrocyte precursor cells are vulnerable to cytokines released under pathological conditions. This review mainly summarizes the effects of cytokines on oligodendrocyte lineage cells in central nervous system diseases. A comprehensive understanding of the effects of cytokines on oligodendrocyte lineage cells contributes to our understanding of central nervous system diseases and offers insights into treatment strategies.
Collapse
Affiliation(s)
- Chengfu Zhang
- Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Mengsheng Qiu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Hui Fu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
19
|
Naseem S, Sun L, Qiu J. Stress granules in atherosclerosis: Insights and therapeutic opportunities. Curr Probl Cardiol 2024; 49:102760. [PMID: 39059785 DOI: 10.1016/j.cpcardiol.2024.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Atherosclerosis, a complex inflammatory and metabolic disorder, is the underlying cause of several life-threatening cardiovascular diseases. Stress granules (SG) are biomolecular condensates composed of proteins and mRNA that form in response to stress. Recent studies suggest a potential link between SG and atherosclerosis development. However, there remain gaps in understanding SG role in atherosclerosis development. Here we provide a thorough analysis of the role of SG in atherosclerosis, covering cellular stresses stimulation, core components, and regulatory genes in SG formation. Furthermore, we explore atherosclerosis induced factors such as inflammation, low or oscillatory shear stress (OSS), and oxidative stress (OS) may impact SG formation and then the development of atherosclerotic lesions. We have assessed how changes in SG dynamics impact pro-atherogenic processes like endothelial dysfunction, lipid metabolism, and immune cell recruitment in atherosclerosis. In summary, this review emphasizes the complex interplay between SG and atherosclerosis that could open innovative directions for targeted therapeutic strategies in preventing or treating atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lijuan Sun
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
20
|
Ortiz Flores RM, Cáceres CS, Cortiñas TI, Gomez Mejiba SE, Sasso CV, Ramirez DC, Mattar Domínguez MA. Exotoxins secreted by Clostridium septicum induce macrophage death: Implications for bacterial immune evasion mechanisms at infection sites. Toxicon 2024; 249:108070. [PMID: 39127083 DOI: 10.1016/j.toxicon.2024.108070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
The induction of macrophage death is considered a potential mechanism by which components secreted by Clostridium septicum are used to evade the innate immune response and cause tissue damage. This study aimed to determine the effects of partially purified fractions of extracellular proteins secreted by C. septicum on the death of mouse peritoneal macrophages. Elicited mouse peritoneal macrophages were incubated with partially purified fractions of proteins secreted by C. septicum into the culture medium. After incubation, the protein fraction with a molecular weight ≥100 kDa caused significant cell death in macrophages, altered cell morphology, increased the expression of markers of apoptosis and autophagy, and increased the expression (protein and mRNA) of IL-10 and TNFα. Our data suggest that the proteins secreted by C. septicum (MW, ≥100 kDa) induce cell death in macrophages by promoting autophagy-triggered apoptosis. This study may contribute to our understanding of the molecular mechanism of immune evasion by C. septicum at the infection site.
Collapse
Affiliation(s)
- R M Ortiz Flores
- Department of Human Physiology, School of Medicine, CAMPUS TEATINOS C/Boulevard Luis Pasteur, University of Malaga, 29010, Malaga, Malaga, Spain.
| | - C S Cáceres
- Laboratory of Microbiology, School of Chemistry Biochemistry and Pharmacy, National University of San Luis, 5700, San Luis, San Luis, Argentina.
| | - T I Cortiñas
- Laboratory of Microbiology, School of Chemistry Biochemistry and Pharmacy, National University of San Luis, 5700, San Luis, San Luis, Argentina.
| | - S E Gomez Mejiba
- Laboratory of Experimental Therapeutics and Nutrition, IMIBIO-SL, CCT-San Luis, CONICET-National University of San Luis, 5700, San Luis, San Luis, Argentina.
| | - C V Sasso
- Department of Medicine and Dermatology, School of Medicine, CAMPUS TEATINOS, C/Boulevard Luis Pasteur, University of Malaga, 29010, Malaga, Malaga, Spain.
| | - D C Ramirez
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL, CCT-San Luis, CONICET-National University of San Luis, 5700, San Luis, San Luis, Argentina.
| | - M A Mattar Domínguez
- Laboratory of Microbiology, School of Chemistry Biochemistry and Pharmacy, National University of San Luis, 5700, San Luis, San Luis, Argentina.
| |
Collapse
|
21
|
Yao J, Zhang L, Zhang C, Chen X, Bao K, Hou S, Yin Y, Liu K, Wen Q, Huang X, Song L. Rhythmic gamma frequency light flickering ameliorates stress-related behaviors and cognitive deficits by modulating neuroinflammatory response through IL-12-Mediated cytokines production in chronic stress-induced mice. Brain Behav Immun 2024; 121:213-228. [PMID: 39043349 DOI: 10.1016/j.bbi.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 06/26/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
Chronic stress enhances the risk for psychiatric disorders and induces depression and cognitive impairment. Gamma oscillations are essential for neurocircuit function, emotion, and cognition. However, the influence of gamma entrainment by sensory stimuli on specific aspects of chronic stress-induced responses remains unclear. Mice were subjected to corticosterone (CORT) administration and chronic restraint stress (CRS) for weeks, followed by rhythmic gamma frequency light flickering exposure. Local field potentials (LFPs) were recorded from the V1, CA1, and PFC regions to verify the light flicker on gamma oscillations. Behavioral tests were used to examine stress-related and memory-related behaviors. Golgi staining was performed to observe changes in spine morphology. Synaptosomes were isolated to determine the expression of synapse-related proteins through immunoblotting. RNA sequencing (RNA-seq) was applied to explore specific changes in the transcriptome. Immunofluorescence staining, real-time quantitative polymerase chain reaction (qPCR), and ELISA were used to evaluate microglial activation and cytokine levels. In this study, we demonstrated that rhythmic 40 Hz LF attenuated stress-related behavior and cognitive impairments by ameliorating the microstructural alterations in spine morphology and increasing the expression of GluN2A and GluA1 in chronically stressed mice. Transcriptome analysis revealed that significantly downregulated genes in LF-exposed CRS mice were enriched in neuroimmune-related signaling pathways. Rhythmic 40 Hz LF exposure significantly decreased the number of Iba1-positive microglia in the PFC and hippocampus, and the expression levels of the M1 markers of microglia iNOS and CD68 were reduced significantly in CRS mice. In addition, 40 Hz LF exposure suppressed the secretion of cytokines IL-12, which could regulate the production of IFN-γ and IL-10 in stressed mice. Our results demonstrate that exposure to rhythmic 40 Hz LF induces the neuroimmune response and downregulation of neuroinflammation with attenuated stress-related behaviors and cognitive function in CRS-induced mice. Our findings highlight the importance of sensory-evoked gamma entrainment as a potential therapeutic strategy for stress-related disorders treatment. Abbreviations: CORT, Chronic corticosterone treatment; CRS, Chronic restraint stress; IACUC, Institutional Animal Care and Use Committee; LF, light flickers; FST, Forced swim test; NSFT, Novelty-suppressed feeding test; SPT, Sucrose preference test; NSFT, Novelty-suppressed feeding; qPCR, Quantitative real-time polymerase chain reaction; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis; PVDF, polyvinylidene fluoride; PBS, phosphate-buffered saline; PBS-T, phosphate-buffered saline plus 0.1% Tween 20; PVDF, polyvinylidene fluoride; GFAP, Glial fibrillary acidic protein; DAPI, 4',6-Diamid- ino-2-phenylindole; Iba1, Ionized calcium-binding adaptor molecule 1; iNOS, Inducible nitric oxide synthase; IL-10, Interleukin-10; IL6, Interleukin 6; IL-1β, Interleukin 1β; IL-12, Interleukin 12; TNF-α, Tumor necrosis factor alpha; IFN-γ, Interferon-gamma; TLR6 and 9, Toll-like Receptor 6 and 9.
Collapse
Affiliation(s)
- Junqi Yao
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China; Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing 100850, China
| | - Chunkui Zhang
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xing Chen
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Ke Bao
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Shaojun Hou
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Yongyu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing 100850, China
| | - Kun Liu
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Qing Wen
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China.
| | - Lun Song
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China.
| |
Collapse
|
22
|
Gohal G, Moni SS, Bakkari MA, Elmobark ME. A Review on Asthma and Allergy: Current Understanding on Molecular Perspectives. J Clin Med 2024; 13:5775. [PMID: 39407835 PMCID: PMC11476424 DOI: 10.3390/jcm13195775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma, a complex disease characterized by persistent airway inflammation, remains an urgent global health concern. We explored the critical role of allergic biomarkers and dysregulated immune system in asthma through an extensive literature review in databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. This review summarizes the growing data on the pivotal role of allergic biomarkers and dysregulated immune system in the development and evolution of asthma. Recent studies have uncovered several biomarkers that elucidate intrinsic allergic mechanisms in individuals with asthma. This article highlights these biomarkers' potential in predicting asthma onset, assessing its intensity, guiding therapeutic interventions, and tracking disease progression. We also explore the innovative therapeutic prospects arising from the convergence of allergy and dysregulated immune system in asthma and emphasize the potential for precision medicine approaches. Understanding allergic biomarkers intertwined with a dysregulated immune system heralds a new era in asthma treatment and points to improved and individualized treatment modalities.
Collapse
Affiliation(s)
- Gassem Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
23
|
Banerjee S, Oguljahan B, Thompson WE, Chowdhury I. Neuregulin 1 Signaling Attenuates Tumor Necrosis Factor α-Induced Female Rat Luteal Cell Death. Endocrinology 2024; 165:bqae129. [PMID: 39312480 PMCID: PMC11456883 DOI: 10.1210/endocr/bqae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/16/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
The corpus luteum (CL) is a transient ovarian endocrine structure that maintains pregnancy in primates during the first trimester and in rodents during the entire pregnancy by producing steroid hormone progesterone (P4). CL lifespan, growth, and differentiation are tightly regulated by survival and cell death signals through luteotrophic and luteolytic factors, including the epidermal growth factor (EGF)-like factor family. Neuregulin 1 (NRG1), a member of the EGF family, mediates its effect through ErbB2/3 receptors. However, the functional role of NRG1 in luteal cells (LCs) is unknown. Thus, this study investigated the role of NRG1 and its molecular mechanism of action in rat LC. Our experimental results suggest a strong positive correlation between steroidogenic acute regulatory protein (StAR) and NRG1 expression in mid-CL and serum P4 and estrogen (E2) production. In contrast, there was a decrease in StAR and NRG1 expression and P4 and E2 production with an increase in tumor necrosis factor α (TNFα) expression in regressing CL. Further in vitro studies in LCs showed that the knockdown of endogenous Nrg1 promoted the expression of proinflammatory and proapoptotic factors and decreased prosurvival factor expression. Subsequently, treatment with exogenous TNFα under these experimental conditions profoundly elevated proinflammatory and proapoptotic factors. Further analysis demonstrated that the phosphorylation status of ErbB2/3, PI3K, Ak strain transforming or protein kinase B (Akt), and ErK1/2 was significantly inhibited under these experimental conditions, whereas the treatment of TNFα further inhibited the phosphorylation of ErbB2/3, PI3K, Akt, and ErK1/2. Collectively, these studies provide new insights into the NRG1-mediated immunomodulatory and prosurvival role in LCs, which may maintain the function of CL.
Collapse
Affiliation(s)
- Saswati Banerjee
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Babayewa Oguljahan
- Center for Laboratory Animal Resources, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Winston E Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Indrajit Chowdhury
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
24
|
Diederich J, Müller KE. Involvement of Root Canal Treatment in Pro-Inflammatory Processes - A Real-World Study. Pragmat Obs Res 2024; 15:165-172. [PMID: 39346572 PMCID: PMC11438457 DOI: 10.2147/por.s479124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
Purpose Endodontic inflammation typically results from root canal infections and sensitizations to filling materials after root canal treatment (RCT), potentially leading to systemic inflammation and disease. We therefore aimed to characterize the inflammatory alterations after RCT as well as the inflammatory molecule levels following tooth extraction or renewed RCT. Patients and Methods All (a total of 2585) walk-in patients with or without RCT history were included in this retrospective study. During the 3-year observation period, blood levels of RANTES/CCL5 (regulated on activation, normal T-cell expressed and secreted/chemotactic cytokine ligand 5), C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and interleukin-10 (IL-10) were measured before and after tooth extraction or renewed RCT. Control patients did not undergo any dental treatment. Results A total 49.38% of patients had a history of at least one RCT. In RCT patients, RANTES/CCL5 values were significantly reduced by both tooth extraction (p=0.03) and renewed dental RCT (p=0.038), while a non-significant increase was observed in untreated patients. TNF-α levels were reduced by tooth extraction (p=0.014) but not by renewed RCT and no intervention. CRP levels were not significantly changed by either treatment. Tooth extraction significantly lowered IFN-γ levels (p=0.003), while in control patients, IFN-γ levels did not change. IL-10 levels were non-significantly reduced by tooth extraction (p=0.061). In a subcohort of RCT patients, the lymphocyte transformation test revealed an allergic reaction to at least one of the root filling materials in 39.46% of patients, with raw gutta percha (56%) and eugenol (19%) being frequent triggers. Conclusion Here, we demonstrate the involvement of root-treated teeth in inflammatory processes, as tooth extraction and renewed RCT could significantly reduce individual cytokine levels. Our data support the use of biomarkers for in vivo monitoring of treatment success.
Collapse
Affiliation(s)
- Joé Diederich
- Center for General, Clinical Environmental and Functional Medicine, Colmar-Berg, Luxembourg
- University of Luxembourg, Faculty of Science, Technology and Medicine, Department of Life Sciences and Medicine, Esch-sur-Alzette, Luxembourg
- Master of Science Program for Preventive and Functional Medicine, Dresden International University, Dresden, Germany
| | - Kurt E Müller
- Master of Science Program for Preventive and Functional Medicine, Dresden International University, Dresden, Germany
| |
Collapse
|
25
|
Yan C, Geng A, Pan Z, Zhang Z, Cui F. MultiFeatVotPIP: a voting-based ensemble learning framework for predicting proinflammatory peptides. Brief Bioinform 2024; 25:bbae505. [PMID: 39406523 PMCID: PMC11479713 DOI: 10.1093/bib/bbae505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/01/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Inflammatory responses may lead to tissue or organ damage, and proinflammatory peptides (PIPs) are signaling peptides that can induce such responses. Many diseases have been redefined as inflammatory diseases. To identify PIPs more efficiently, we expanded the dataset and designed an ensemble learning model with manually encoded features. Specifically, we adopted a more comprehensive feature encoding method and considered the actual impact of certain features to filter them. Identification and prediction of PIPs were performed using an ensemble learning model based on five different classifiers. The results show that the model's sensitivity, specificity, accuracy, and Matthews correlation coefficient are all higher than those of the state-of-the-art models. We named this model MultiFeatVotPIP, and both the model and the data can be accessed publicly at https://github.com/ChaoruiYan019/MultiFeatVotPIP. Additionally, we have developed a user-friendly web interface for users, which can be accessed at http://www.bioai-lab.com/MultiFeatVotPIP.
Collapse
Affiliation(s)
- Chaorui Yan
- School of Computer Science and Technology, Hainan University, 58 Renmin Avenue, Meilan District, Haidian Campus, Haikou 570228, China
| | - Aoyun Geng
- School of Computer Science and Technology, Hainan University, 58 Renmin Avenue, Meilan District, Haidian Campus, Haikou 570228, China
| | - Zhuoyu Pan
- International Business School, Hainan University, 58 Renmin Avenue, Meilan District, Haidian Campus, Haikou 570228, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, 58 Renmin Avenue, Meilan District, Haidian Campus, Haikou 570228, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, 58 Renmin Avenue, Meilan District, Haidian Campus, Haikou 570228, China
| |
Collapse
|
26
|
Nasr S, Dawood AS, Ibrahim AM, Abdel-Aziz MS, Fayad W, Abdelnaser A, El-Hady FKA. Anti-inflammatory potential of aspergillus unguis SP51-EGY: TLR4-dependent effects & chemical diversity via Q-TOF LC-HRMS. BMC Biotechnol 2024; 24:62. [PMID: 39294631 PMCID: PMC11411751 DOI: 10.1186/s12896-024-00890-1] [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: 10/06/2023] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
Inflammation serves as an intricate defense mechanism for tissue repair. However, overactivation of TLR4-mediated inflammation by lipopolysaccharide (LPS) can lead to detrimental outcomes such as sepsis, acute lung injury, and chronic inflammation, often associated with cancer and autoimmune diseases. This study delves into the anti-inflammatory properties of "Aspergillus unguis isolate SP51-EGY" on LPS-stimulated RAW 264.7 macrophages. Through real-time qPCR, we assessed the expression levels of pivotal inflammatory genes, including iNOS, COX-2, TNF-α, and IL-6. Remarkably, our fungal extracts significantly diminished NO production and showed noteworthy reductions in the mRNA expression levels of the aforementioned genes. Furthermore, while Nrf2 is typically associated with modulating inflammatory responses, our findings indicate that the anti-inflammatory effects of our extracts are not Nrf2-dependent. Moreover, the chemical diversity of the potent extract (B Sh F) was elucidated using Q-TOF LC-HRMS, identifying 54 compounds, some of which played vital roles in suppressing inflammation. Most notably, compounds like granisetron, fenofibrate, and umbelliprenin were found to downregulate TNF-α, IL-1β, and IL-6 through the NF-κB signaling pathway. In conclusion, "Aspergillus unguis isolate SP51-EGY", isolated from the Red Sea, Egypt, has been unveiled as a promising TLR4 inhibitor with significant anti-inflammatory potentials, presenting novel insights for their potential therapeutic use in inflammation.
Collapse
Affiliation(s)
- Soad Nasr
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt
- Biochemical Engineering Department, Faculty of Energy and Environmental Engineering, The British University in Egypt, Suez Desert Road, P.O. Box: 43, El-Shorouk City, Cairo, 11837, Egypt
| | - Abdelhameed S Dawood
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt
| | - Amal Mosad Ibrahim
- Chemistry of Natural and Microbial Products Department, National Research Centre, Giza, 12622, Egypt
| | | | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Giza, 12622, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt.
| | - Faten K Abd El-Hady
- Chemistry of Natural and Microbial Products Department, National Research Centre, Giza, 12622, Egypt
| |
Collapse
|
27
|
Kraski A, Migdał P, Klopfleisch R, Räckel C, Sharbati J, Heimesaat MM, Alter T, Hanisch C, Gölz G, Einspanier R, Sharbati S. Structured multicellular intestinal spheroids (SMIS) as a standardized model for infection biology. Gut Pathog 2024; 16:47. [PMID: 39289703 PMCID: PMC11406839 DOI: 10.1186/s13099-024-00644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND 3D cell culture models have recently garnered increasing attention for replicating organ microarchitecture and eliciting in vivo-like responses, holding significant promise across various biological disciplines. Broadly, 3D cell culture encompasses organoids as well as single- and multicellular spheroids. While the latter have found successful applications in tumor research, there is a notable scarcity of standardized intestinal models for infection biology that mimic the microarchitecture of the intestine. Hence, this study aimed to develop structured multicellular intestinal spheroids (SMIS) specifically tailored for studying molecular basis of infection by intestinal pathogens. RESULTS We have successfully engineered human SMIS comprising four relevant cell types, featuring a fibroblast core enveloped by an outer monolayer of enterocytes and goblet cells along with monocytic cells. These SMIS effectively emulate the in vivo architecture of the intestinal mucosal surface and manifest differentiated morphological characteristics, including the presence of microvilli, within a mere two days of culture. Through analysis of various differentiation factors, we have illustrated that these spheroids attain heightened levels of differentiation compared to 2D monolayers. Moreover, SMIS serve as an optimized intestinal infection model, surpassing the capabilities of traditional 2D cultures, and exhibit a regulatory pattern of immunological markers similar to in vivo infections after Campylobacter jejuni infection. Notably, our protocol extends beyond human spheroids, demonstrating adaptability to other species such as mice and pigs. CONCLUSION Based on the rapid attainment of enhanced differentiation states, coupled with the emergence of functional brush border features, increased cellular complexity, and replication of the intestinal mucosal microarchitecture, which allows for exposure studies via the medium, we are confident that our innovative SMIS model surpasses conventional cell culture methods as a superior model. Moreover, it offers advantages over stem cell-derived organoids due to scalability and standardization capabilities of the protocol. By showcasing differentiated morphological attributes, our model provides an optimal platform for diverse applications. Furthermore, the investigated differences of several immunological factors compared to monotypic monolayers after Campylobacter jejuni infection underline the refinement of our spheroid model, which closely mimics important features of in vivo infections.
Collapse
Affiliation(s)
- Angelina Kraski
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Paweł Migdał
- Institute of Animal Husbandry and Breeding, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Clara Räckel
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | | | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Demeter F, Bihari G, Vadicsku D, Sinkovits G, Kajdácsi E, Horváth L, Réti M, Müller V, Iványi Z, Gál J, Gopcsa L, Reményi P, Szathmáry B, Lakatos B, Szlávik J, Bobek I, Prohászka ZZ, Förhécz Z, Masszi T, Vályi-Nagy I, Prohászka Z, Cervenak L. Anti-Inflammatory Cytokine Profiles in Thrombotic Thrombocytopenic Purpura-Differences Compared to COVID-19. Int J Mol Sci 2024; 25:10007. [PMID: 39337495 PMCID: PMC11432022 DOI: 10.3390/ijms251810007] [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: 08/13/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Thromboinflammation/immunothrombosis plays a role in several diseases including thrombotic thrombocytopenic purpura (TTP) and COVID-19. Unlike the extensive research that has been conducted on COVID-19 cytokine storms, the baseline and acute phase cytokine profiles of TTP are poorly characterized. Moreover, we compared the cytokine profiles of TTP and COVID-19 to identify the disease-specific/general characteristics of thromboinflammation/immunothrombosis. Plasma concentrations of 33 soluble mediators (SMs: cytokines, chemokines, soluble receptors, and growth factors) were measured by multiplex bead-based LEGENDplex™ immunoassay from 32 COVID-19 patients (32 non-vaccinated patients in three severity groups), 32 TTP patients (remission/acute phase pairs of 16 patients), and 15 control samples. Mainly, the levels of innate immunity-related SMs changed in both diseases. In TTP, ten SMs decreased in both remission and acute phases compared to the control, one decreased, and two increased only in the acute phase compared to remission, indicating mostly anti-inflammatory changes. In COVID-19, ten pro-inflammatory SMs increased, whereas one decreased with increasing severity compared to the control. In severe COVID-19, sixteen SMs exceeded acute TTP levels, with only one higher in TTP. PCA identified CXCL10, IL-1RA, and VEGF as the main discriminators among their cytokine profiles. The innate immune response is altered in both diseases. The cytokine profile of TTP suggests a distinct pathomechanism from COVID-19 and supports referring to TTP as thromboinflammatory rather than immunothrombotic, emphasizing thrombosis over inflammation as the driving force of the acute phase.
Collapse
Affiliation(s)
- Flóra Demeter
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Bihari
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Dorina Vadicsku
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Sinkovits
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - Laura Horváth
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Marienn Réti
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Zsolt Iványi
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - János Gál
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - László Gopcsa
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Péter Reményi
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Beáta Szathmáry
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Botond Lakatos
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - János Szlávik
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Ilona Bobek
- Department of Anaesthesiology and Intensive Therapy, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zita Z. Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Zsolt Förhécz
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Tamás Masszi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - István Vályi-Nagy
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
29
|
Cheng Z, Liu B, Liu X. Circadian gene signatures in the progression of obesity based on machine learning and Mendelian randomization analysis. Front Nutr 2024; 11:1407265. [PMID: 39351493 PMCID: PMC11439728 DOI: 10.3389/fnut.2024.1407265] [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: 03/26/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Obesity, a global health concern, is associated with a spectrum of chronic diseases and cancers. Our research sheds light on the regulatory role of circadian genes in obesity progression, providing insight into the immune landscape of obese patients, and introducing new avenues for therapeutic interventions. Methods Expression files of multiple datasets were retrieved from the GEO database. By 80 machine-learning algorithm combinations and Mendelian randomization analysis, we discovered the key circadian genes contributing to and protecting against obesity. Subsequently, an immune infiltration analysis was conducted to examine the alterations in immune cell types and their abundance in the body and to investigate the relationships between circadian genes and immune cells. Furthermore, we delved into the molecular mechanisms of key genes implicated in obesity. Results Our study identified three key circadian genes (BHLHE40, PPP1CB, and CSNK1E) associated with obesity. BHLHE40 was found to promote obesity through various pathways, while PPP1CB and CSNK1E counteracted lipid metabolism disorders, and modulated cytokines, immune receptors, T cells, and monocytes. Conclusion In conclusion, the key circadian genes (BHLHE40, CSNK1E, and PPP1CB) may serve as novel biomarkers for understanding obesity pathogenesis and have significant correlations with infiltrating immune cells, thus providing potential new targets for obese prevention and treatment.
Collapse
Affiliation(s)
- Zhi’ang Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Binghong Liu
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Xiaoyong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Ophthalmology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| |
Collapse
|
30
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [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: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
31
|
Ma X, Wang WX. Unveiling osmoregulation and immunological adaptations in Eleutheronema tetradactylum gills through high-throughput single-cell transcriptome sequencing. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109878. [PMID: 39245186 DOI: 10.1016/j.fsi.2024.109878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
The fourfinger threadfin fish (Eleutheronema tetradactylum) is an economically significant species renowned for its ability to adapt to varying salinity environments, with gills serving as their primary organs for osmoregulation and immune defense. Previous studies focused on tissue and morphological levels, whereas ignored the cellular heterogeneity and the crucial gene information related to core cell subsets within E. tetradactylum gills. In this study, we utilized high-throughput single-cell RNA sequencing (scRNA-seq) to analyze the gills of E. tetradactylum, characterizing 16 distinct cell types and identifying unique gene markers and enriched functions associated within each cell type. Additionally, we subdivided ionocyte cells into four distinct subpopulations for the first time in E. tetradactylum gills. By employing weighted gene co-expression network analysis (WGCNA), we further investigated the cellular heterogeneity and specific response mechanisms to salinity fluctuant. Our findings revealed the intricate osmoregulation and immune functions of gill cells, highlighting their crucial roles in maintaining homeostasis and adapting to fluctuating salinity levels. This comprehensive cell-type atlas provides valuable insights into the species adaptive strategies, contributing to the conservation and management of this commercially significant fish as well as other euryhaline species.
Collapse
Affiliation(s)
- Xiaoli Ma
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China.
| |
Collapse
|
32
|
Rinker DC, Sauters TJC, Steffen K, Gumilang A, Raja HA, Rangel-Grimaldo M, Pinzan CF, de Castro PA, Dos Reis TF, Delbaje E, Houbraken J, Goldman GH, Oberlies NH, Rokas A. Strain heterogeneity in a non-pathogenic Aspergillus fungus highlights factors associated with virulence. Commun Biol 2024; 7:1082. [PMID: 39232082 PMCID: PMC11374809 DOI: 10.1038/s42003-024-06756-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Fungal pathogens exhibit extensive strain heterogeneity, including variation in virulence. Whether closely related non-pathogenic species also exhibit strain heterogeneity remains unknown. Here, we comprehensively characterized the pathogenic potentials (i.e., the ability to cause morbidity and mortality) of 16 diverse strains of Aspergillus fischeri, a non-pathogenic close relative of the major pathogen Aspergillus fumigatus. In vitro immune response assays and in vivo virulence assays using a mouse model of pulmonary aspergillosis showed that A. fischeri strains varied widely in their pathogenic potential. Furthermore, pangenome analyses suggest that A. fischeri genomic and phenotypic diversity is even greater. Genomic, transcriptomic, and metabolic profiling identified several pathways and secondary metabolites associated with variation in virulence. Notably, strain virulence was associated with the simultaneous presence of the secondary metabolites hexadehydroastechrome and gliotoxin. We submit that examining the pathogenic potentials of non-pathogenic close relatives is key for understanding the origins of fungal pathogenicity.
Collapse
Affiliation(s)
- David C Rinker
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Thomas J C Sauters
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Karin Steffen
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Adiyantara Gumilang
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Huzefa A Raja
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Manuel Rangel-Grimaldo
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Camila Figueiredo Pinzan
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Patrícia Alves de Castro
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Thaila Fernanda Dos Reis
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Endrews Delbaje
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Jos Houbraken
- Food and Indoor Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Gustavo H Goldman
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil.
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA.
| | - Antonis Rokas
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
33
|
Coperchini F, Greco A, Teliti M, Croce L, Chytiris S, Magri F, Gaetano C, Rotondi M. Inflamm-ageing: How cytokines and nutrition shape the trajectory of ageing. Cytokine Growth Factor Rev 2024:S1359-6101(24)00065-0. [PMID: 39237438 DOI: 10.1016/j.cytogfr.2024.08.004] [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: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Population ageing is increasing in prevalence in most developed countries. Ageing is the decline of functional properties at the cellular, tissue, and organ level. Biochemical changes that occur in all organisms that experience biological ageing are referred to as the "Hallmarks of ageing". Inflammation is a common denominator of the hallmarks of ageing, being mechanistically involved in most age-related health consequences. Inflamm-ageing refers to age-related changes in the inflammatory and immune systems which somehow drive the ageing process towards healthy or unhealthy ageing. Current evidences, support that, reversing the age-related pro-inflammatory status of inflamm-ageing, is able to modulate most hallmarks of ageing. Inflamm-ageing is associated with increased levels of pro-inflammatory molecules (e.g. cytokines, chemokines), ultimately producing a chronic low-grade inflammatory state typically observed in older individuals. It is commonly accepted that, the balance between pro- and anti-inflammatory cytokines/chemokines is one of the factors determining whether healthy or unhealthy ageing occurs. Malnutrition and nutritional imbalances, are highly prevalent in the elderly, playing a role in driving the balance of pro- and anti-inflammatory immunoactive molecules. In particular, malnutrition is a major risk factor for sarcopenia, a phenomenon characterized by loss of muscle mass, which is often referred to as the biological basis for frailty. Given the close relationship between malnutrition and sarcopenia, there is also evidence for a link between malnutrition and frailty. Indeed, changes in cytokine/chemokine levels in elderly patients with malnutrition were demonstrated. The demonstration that specific cytokines play a role in modulating appetite and nutrient sensing and taste reception, provided further evidence for the existence of a link between inflamm-ageing, nutrition and cytokines in shaping the trajectory of ageing. The present review will overview current evidence supporting the role of specific circulating cytokines and chemokines in the relationship between ageing, inflammation, and malnutrition.
Collapse
Affiliation(s)
- Francesca Coperchini
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Alessia Greco
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Marsida Teliti
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Laura Croce
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Spyridon Chytiris
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Flavia Magri
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Mario Rotondi
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy.
| |
Collapse
|
34
|
Dhar P, Sominsky L, O'Hely M, Dawson S, Collier F, Tang MLK, Mansell T, Burgner D, Smith C, Hyde N, Downing K, Hesketh KD, Ponsonby AL, Vuillermin P. Physical activity and circulating inflammatory markers and cytokines during pregnancy: A population-based cohort study. Acta Obstet Gynecol Scand 2024; 103:1808-1819. [PMID: 38924074 PMCID: PMC11324931 DOI: 10.1111/aogs.14870] [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: 10/19/2023] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Physical activity (PA) during pregnancy has numerous benefits, which may be mediated via effects on the immune system. However, supportive evidence is inconsistent and is mainly from studies in high-risk groups. We estimated the effect of PA during pregnancy on systemic inflammatory markers and cytokines in mothers recruited in the Barwon infant study. MATERIAL AND METHODS The Barwon infant study is a prebirth cohort of 1064 mothers recruited in the Barwon Region of Victoria, Australia. Participants reported their previous week's PA at their 28-week antenatal appointment using the International PA Questionnaire. Women were grouped into low, moderate, and high PA categories based on daily duration and weekly frequency of walking, moderate- or vigorous-intensity PA. Women reporting moderate levels of PA, consistent with current recommendations, served as the comparison group. Markers of systemic inflammation, high-sensitivity C-reactive protein (hsCRP), glycoprotein acetyls (GlycA), and 17 cytokines were measured at 28 weeks gestation and log transformed as appropriate. Regression analyses adjusted for maternal smoking, gestational diabetes mellitus, prepregnancy BMI, and household size were performed. RESULTS Compared to women in the moderate group (n = 371, 42%), women reporting low PA (n = 436, 50%) had 10.1% higher hsCRP (95% CI (3.7% to 16.6%), p < 0.01) while women in high PA (n = 76, 9%) had a 14% higher hsCRP (95% CI (3.1% to 24.8%), p = 0.01). Women in the high PA category had higher interleukin (IL)-4 (q = 0.03) and IL-9 (q = 0.03) levels compared to those in moderate category. Each vigorous MET minute/week was associated with lower GlycA (β = -0.004, 95% CI (-0.044 to 0.035); p = 0.03). CONCLUSIONS Low and high PA are each associated with higher hsCRP than moderate PA, suggesting that undertaking the recommended moderate PA during pregnancy decreases systemic inflammation. High PA affects T cell-associated cytokines during pregnancy. Evidence from our study suggests that PA can modulate the immune responses during pregnancy. Studies are now required to assess whether PA during pregnancy impacts maternal and infant clinical outcomes by modifying inflammatory responses.
Collapse
Affiliation(s)
- Poshmaal Dhar
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Luba Sominsky
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
- Barwon Health, Geelong, Victoria, Australia
| | - Martin O'Hely
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Samantha Dawson
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Fiona Collier
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Mimi L K Tang
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Craig Smith
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Natalie Hyde
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Katherine Downing
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Kylie D Hesketh
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Peter Vuillermin
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
- Barwon Health, Geelong, Victoria, Australia
| |
Collapse
|
35
|
Kadier K, Niu T, Ding B, Chen B, Qi X, Chen D, Cheng X, Fang Y, Zhou J, Zhao W, Liu Z, Yuan Y, Zhou Z, Dong X, Yang B, He Q, Cao J, Jiang L, Zhu CL. PROTAC-Mediated HDAC7 Protein Degradation Unveils Its Deacetylase-Independent Proinflammatory Function in Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309459. [PMID: 39049738 PMCID: PMC11423193 DOI: 10.1002/advs.202309459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/25/2024] [Indexed: 07/27/2024]
Abstract
Class IIa histone deacetylases (Class IIa HDACs) play critical roles in regulating essential cellular metabolism and inflammatory pathways. However, dissecting the specific roles of each class IIa HDAC isoform is hindered by the pan-inhibitory effect of current inhibitors and a lack of tools to probe their functions beyond epigenetic regulation. In this study, a novel PROTAC-based compound B4 is developed, which selectively targets and degrades HDAC7, resulting in the effective attenuation of a specific set of proinflammatory cytokines in both lipopolysaccharide (LPS)-stimulated macrophages and a mouse model. By employing B4 as a molecular probe, evidence is found for a previously explored role of HDAC7 that surpasses its deacetylase function, suggesting broader implications in inflammatory processes. Mechanistic investigations reveal the critical involvement of HDAC7 in the Toll-like receptor 4 (TLR4) signaling pathway by directly interacting with the TNF receptor-associated factor 6 and TGFβ-activated kinase 1 (TRAF6-TAK1) complex, thereby initiating the activation of the downstream mitogen-activated protein kinase/nuclear factor-κB (MAPK/NF-κB) signaling cascade and subsequent gene transcription. This study expands the insight into HDAC7's role within intricate inflammatory networks and highlights its therapeutic potential as a novel target for anti-inflammatory treatments.
Collapse
Affiliation(s)
- Kailibinuer Kadier
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Tian Niu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Baoli Ding
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Boya Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xuxin Qi
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Danni Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xirui Cheng
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yizheng Fang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jiahao Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Wenyi Zhao
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Zeqi Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yi Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Zhan Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Xiaowu Dong
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, P. R. China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, 310058, P. R. China
| | - Ji Cao
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
| | - Li Jiang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Cheng-Liang Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
36
|
Bhol NK, Bhanjadeo MM, Singh AK, Dash UC, Ojha RR, Majhi S, Duttaroy AK, Jena AB. The interplay between cytokines, inflammation, and antioxidants: mechanistic insights and therapeutic potentials of various antioxidants and anti-cytokine compounds. Biomed Pharmacother 2024; 178:117177. [PMID: 39053423 DOI: 10.1016/j.biopha.2024.117177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Cytokines regulate immune responses essential for maintaining immune homeostasis, as deregulated cytokine signaling can lead to detrimental outcomes, including inflammatory disorders. The antioxidants emerge as promising therapeutic agents because they mitigate oxidative stress and modulate inflammatory pathways. Antioxidants can potentially ameliorate inflammation-related disorders by counteracting excessive cytokine-mediated inflammatory responses. A comprehensive understanding of cytokine-mediated inflammatory pathways and the interplay with antioxidants is paramount for developing natural therapeutic agents targeting inflammation-related disorders and helping to improve clinical outcomes and enhance the quality of life for patients. Among these antioxidants, curcumin, vitamin C, vitamin D, propolis, allicin, and cinnamaldehyde have garnered attention for their anti-inflammatory properties and potential therapeutic benefits. This review highlights the interrelationship between cytokines-mediated disorders in various diseases and therapeutic approaches involving antioxidants.
Collapse
Affiliation(s)
- Nitish Kumar Bhol
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | | | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Umesh Chandra Dash
- Environmental Biotechnology Laboratory, KIIT School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
| | - Rakesh Ranjan Ojha
- Department of Bioinformatics, BJB (A) College, Bhubaneswar, Odisha-751014, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India.
| |
Collapse
|
37
|
Suh YJ, Li AT, Pandey M, Nordmann CS, Huang YL, Wu M. Decoding physical principles of cell migration under controlled environment using microfluidics. BIOPHYSICS REVIEWS 2024; 5:031302. [PMID: 39091432 PMCID: PMC11290890 DOI: 10.1063/5.0199161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
Living cells can perform incredible tasks that man-made micro/nano-sized robots have not yet been able to accomplish. One example is that white blood cells can sense and move to the site of pathogen attack within minutes. The robustness and precision of cellular functions have been perfected through billions of years of evolution. In this context, we ask the question whether cells follow a set of physical principles to sense, adapt, and migrate. Microfluidics has emerged as an enabling technology for recreating well-defined cellular environment for cell migration studies, and its ability to follow single cell dynamics allows for the results to be amenable for theoretical modeling. In this review, we focus on the development of microfluidic platforms for recreating cellular biophysical (e.g., mechanical stress) and biochemical (e.g., nutrients and cytokines) environments for cell migration studies in 3D. We summarize the basic principles that cells (including bacteria, algal, and mammalian cells) use to respond to chemical gradients learned from microfluidic systems. We also discuss about novel biological insights gained from studies of cell migration under biophysical cues and the need for further quantitative studies of cell function under well-controlled biophysical environments in the future.
Collapse
Affiliation(s)
- Young Joon Suh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Alan T. Li
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mrinal Pandey
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Cassidy S. Nordmann
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
38
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
39
|
Naidoo SJ, Naicker T. The Enigmatic Interplay of Interleukin-10 in the Synergy of HIV Infection Comorbid with Preeclampsia. Int J Mol Sci 2024; 25:9434. [PMID: 39273381 PMCID: PMC11395227 DOI: 10.3390/ijms25179434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Cytokines coordinate the intricate choreography of the immune system, directing cellular activities that mediate inflammation, pathogen defense, pathology and tissue repair. Within this spectrum, the anti-inflammatory prowess of interleukin-10 (IL-10) predominates in immune homeostasis. In normal pregnancy, the dynamic shift of IL-10 across trimesters maintains maternal immune tolerance ensuring fetal development and pregnancy success. Unravelling the dysregulation of IL-10 in pregnancy complications is vital, particularly in the heightened inflammatory condition of preeclampsia. Of note, a reduction in IL-10 levels contributes to endothelial dysfunction. In human immunodeficiency virus (HIV) infection, a complex interplay of IL-10 occurs, displaying a paradoxical paradigm of being immune-protective yet aiding viral persistence. Genetic variations in the IL-10 gene further modulate susceptibility to HIV infection and preeclampsia, albeit with nuanced effects across populations. This review outlines the conceptual framework underlying the role of IL-10 in the duality of normal pregnancy and preeclampsia together with HIV infection, thus highlighting its regulatory mechanisms and genetic influences. Synthesizing these findings in immune modulation presents avenues for therapeutic interventions in pregnancy complications comorbid with HIV infection.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Department of Optics and Imaging, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
40
|
Wieg L, Ciola JC, Wasén CC, Gaba F, Colletti BR, Schroeder MK, Hinshaw RG, Ekwudo MN, Holtzman DM, Saito T, Sasaguri H, Saido TC, Cox LM, Lemere CA. Cognitive Effects of Simulated Galactic Cosmic Radiation Are Mediated by ApoE Status, Sex, and Environment in APP Knock-In Mice. Int J Mol Sci 2024; 25:9379. [PMID: 39273325 PMCID: PMC11394682 DOI: 10.3390/ijms25179379] [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: 08/15/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Cosmic radiation experienced during space travel may increase the risk of cognitive impairment. While simulated galactic cosmic radiation (GCRsim) has led to memory deficits in wildtype (WT) mice, it has not been investigated whether GCRsim in combination with genetic risk factors for Alzheimer's disease (AD) worsens memory further in aging mice. Here, we investigated the central nervous system (CNS) effects of 0 Gy (sham) or 0.75 Gy five-ion GCRsim or 2 Gy gamma radiation (IRR) in 14-month-old female and male APPNL-F/NL-F knock-in (KI) mice bearing humanized ApoE3 or ApoE4 (APP;E3F and APP;E4F). As travel to a specialized facility was required for irradiation, both traveled sham-irradiated C57BL/6J WT and KI mice and non-traveled (NT) KI mice acted as controls for potential effects of travel. Mice underwent four behavioral tests at 20 months of age and were euthanized for pathological and biochemical analyses 1 month later. Fecal samples were collected pre- and post-irradiation at four different time points. GCRsim seemed to impair memory in male APP;E3F mice compared to their sham counterparts. Travel tended to improve cognition in male APP;E3F mice and lowered total Aβ in female and male APP;E3F mice compared to their non-traveled counterparts. Sham-irradiated male APP;E4F mice accumulated more fibrillar amyloid than their APP;E3F counterparts. Radiation exposure had only modest effects on behavior and brain changes, but travel-, sex-, and genotype-specific effects were seen. Irradiated mice had immediate and long-term differences in their gut bacterial composition that correlated to Alzheimer's disease phenotypes.
Collapse
Affiliation(s)
- Laura Wieg
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jason C Ciola
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Caroline C Wasén
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Fidelia Gaba
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Brianna R Colletti
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Maren K Schroeder
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert G Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Millicent N Ekwudo
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya 467-8601, Aichi, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan
| | - Laura M Cox
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Chen X, Dai Y, Li Y, Xin J, Zou J, Wang R, Zhang H, Liu Z. Identification of cross-talk pathways and PANoptosis-related genes in periodontitis and Alzheimer's disease by bioinformatics analysis and machine learning. Front Aging Neurosci 2024; 16:1430290. [PMID: 39258145 PMCID: PMC11384588 DOI: 10.3389/fnagi.2024.1430290] [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: 05/09/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Background and objectives Periodontitis (PD), a chronic inflammatory disease, is a serious threat to oral health and is one of the risk factors for Alzheimer's disease (AD). A growing body of evidence suggests that the two diseases are closely related. However, current studies have not provided a comprehensive understanding of the common genes and common mechanisms between PD and AD. This study aimed to screen the crosstalk genes of PD and AD and the potential relationship between cross-talk and PANoptosis-related genes. The relationship between core genes and immune cells will be analyzed to provide new targets for clinical treatment. Materials and methods The PD and AD datasets were downloaded from the GEO database and differential expression analysis was performed to obtain DEGs. Overlapping DEGs had cross-talk genes linking PD and OP, and PANoptosis-related genes were obtained from a literature review. Pearson coefficients were used to compute cross-talk and PANoptosis-related gene correlations in the PD and AD datasets. Cross-talk genes were obtained from the intersection of PD and AD-related genes, protein-protein interaction(PPI) networks were constructed and cross-talk genes were identified using the STRING database. The intersection of cross-talk and PANoptosis-related genes was defined as cross-talk-PANoptosis genes. Core genes were screened using ROC analysis and XGBoost. PPI subnetwork, gene-biological process, and gene-pathway networks were constructed based on the core genes. In addition, immune infiltration on the PD and AD datasets was analyzed using the CIBERSORT algorithm. Results 366 cross-talk genes were overlapping between PD DEGs and AD DEGs. The intersection of cross-talk genes with 109 PANoptosis-related genes was defined as cross-talk-PANoptosis genes. ROC and XGBoost showed that MLKL, DCN, IL1B, and IL18 were more accurate than the other cross-talk-PANoptosis genes in predicting the disease, as well as better in overall characterization. GO and KEGG analyses showed that the four core genes were involved in immunity and inflammation in the organism. Immune infiltration analysis showed that B cells naive, Plasma cells, and T cells gamma delta were significantly differentially expressed in patients with PD and AD compared with the normal group. Finally, 10 drugs associated with core genes were retrieved from the DGIDB database. Conclusion This study reveals the joint mechanism between PD and AD associated with PANoptosis. Analyzing the four core genes and immune cells may provide new therapeutic directions for the pathogenesis of PD combined with AD.
Collapse
Affiliation(s)
- Xiantao Chen
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yifei Dai
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yushen Li
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Jiajun Xin
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Jiatong Zou
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Rui Wang
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Hao Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Zhihui Liu
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| |
Collapse
|
42
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
43
|
Mohan S, Krishnan L, Madhusoodanan N, Sobha A, Babysulochana AD, Vankadari N, Purushothaman J, Somappa SB. Ligand-Based Pharmacophoric Design and Anti-inflammatory Evaluation of Triazole Linked Semisynthetic Labdane Conjugates. ACS Med Chem Lett 2024; 15:1260-1268. [PMID: 39140047 PMCID: PMC11318007 DOI: 10.1021/acsmedchemlett.4c00141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
This study employed a ligand-based pharmacophoric approach to design and synthesize 33 novel semisynthetic labdane-appended triazolyl isatins to discover potential anti-inflammatory agents. The anti-inflammatory efficacy of the derivatives was evaluated by their ability to inhibit the production of NO, TNF-α, and IL-6, in lipopolysaccharide-induced RAW264.7 macrophages. The initial screening revealed that compound 7a ((1-(2-(2,3-dioxoindolin-1-yl)ethyl)-1H-1,2,3-triazol-4-yl)methyl (E)-3-formyl-5-((1S,4aS,8aS)-5,5,8a-trimethyl-2-methylenedecahydronaphthalen-1-yl)pent-3-enoate) exhibited an anti-inflammatory effect (NO inhibition, IC50 = 3.13 μΜ), surpassing both the positive control indomethacin (NO inhibition, IC50 = 7.31 μΜ) and the parent compound labdane dialdehyde. Notably, 7a reduced the levels of pro-inflammatory cytokines TNF-α and IL-6 while increasing the levels of the anti-inflammatory cytokine IL-10. Mechanistic studies revealed that 7a downregulated the expression of COX-2 and iNOS by inhibiting the NF-κB signaling pathway. In silico molecular modeling studies on NF-κB proteins support these findings, suggesting that 7a is a promising candidate for developing into a potent anti-inflammatory clinical agent.
Collapse
Affiliation(s)
- Sangeetha Mohan
- Chemical
Sciences and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Lekshmy Krishnan
- Agro
Processing and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
| | - Nithya Madhusoodanan
- Chemical
Sciences and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Anjali Sobha
- Chemical
Sciences and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Alansheeja D. Babysulochana
- Chemical
Sciences and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Department
of Chemistry, Government Arts College, Thiruvananthapuram, Kerala 695 014, India
| | - Naveen Vankadari
- Department
of Biochemistry and Pharmacology, Bio21 Institute, The University of Melbourne, Melbourne, Victoria VIC 3052, Australia
| | - Jayamurthy Purushothaman
- Agro
Processing and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Sasidhar B. Somappa
- Chemical
Sciences and Technology Division, CSIR−National
Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695
019, Kerala India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| |
Collapse
|
44
|
Wu Y, Huang JY, Conlon MT, Shenoy MK, Chao JL, Chooi MY, Koch MA, Gerner MY. Distal Immunization and Systemic Cytokines Establish a Transient Immune Alert State in the Intestine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:373-383. [PMID: 38884660 PMCID: PMC11250722 DOI: 10.4049/jimmunol.2400209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024]
Abstract
Conventionally, immune responses are studied in the context of inflamed tissues and their corresponding draining lymph nodes (LNs). However, little is known about the effects of systemic inflammatory signals generated during local inflammation on distal tissues and nondraining LNs. Using a mouse model of cutaneous immunization, we found that systemic inflammatory stimuli triggered a rapid and selective distal response in the small intestine and the mesenteric LN (mesLN). This consisted of increased permeability of intestinal blood vessels and lymphatic drainage of bloodborne solutes into the mesLN, enhanced activation and migration of intestinal dendritic cells, as well as amplified T cell responses in the mesLNs to systemic but not orally derived Ags. Mechanistically, we found that the small intestine endothelial cells preferentially expressed molecules involved in TNF-α signaling and that TNF-α blockade markedly diminished distal intestinal responses to cutaneous immunization. Together, these findings reveal that the intestinal immune system is rapidly and selectively activated in response to inflammatory cues regardless of their origin, thus identifying an additional layer of defense and enhanced surveillance of a key barrier organ at constant risk of pathogen encounter.
Collapse
Affiliation(s)
- Yixuan Wu
- Department of Immunology, University of Washington, Seattle, WA
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Jessica Y Huang
- Department of Immunology, University of Washington, Seattle, WA
| | | | - Meera K Shenoy
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jaime L Chao
- Department of Immunology, University of Washington, Seattle, WA
| | - Ming Yao Chooi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Meghan A Koch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | |
Collapse
|
45
|
Liu Y, Song Y, Cheng X, Guo X, Yun S, Lu Y, Li M, Wang J, Zou J. A cytokine-like factor 1 homolog acts as a macrophage chemoattractant in grass carp. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109722. [PMID: 38925447 DOI: 10.1016/j.fsi.2024.109722] [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: 04/04/2024] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Cytokine-like factor 1 (CYTL1) is a small cytokine and has diverse biological functions in mammals. However, whether CYTL1 exists in lower vertebrates is not clear. In this study, we identified cytl homologs in fish and characterized the immune functions in a teleost species, grass carp (Ctenopharyngodon idella). Fish CYTL1 homologs share conserved molecular features with their mammalian counterparts, including 6 cysteine residues in the mature peptide, genomic organization and synteny. Gene expression analysis revealed that cytl1 was constitutively expressed in tissues of grass carp, with the highest expression detected in the heart. Upon infection with Aeromonas hydrophila (A. hydrophila), cytl1 was downregulated in the hindgut, head kidney, skin, and spleen. In the primary head kidney leukocytes (HKLs), stimulation with inactivated A. hydrophila, LPS, poly(I:C), IL-22, IFN-a or IFN-γrel resulted in downregulation of cytl1 expression. Recombinant grass carp CYTL1 protein produced in the HEK293-F cells was potent to induce il-10 expression, but had little effect on the expression of il-1β and il-6. In vivo experiments revealed that CYTL1 was effective to recruit macrophages to the muscle injected with cytl expression plasmids. Taken together, our results indicate that CYTL1 is a potent chemokine for recruitment of macrophages in fish.
Collapse
Affiliation(s)
- Yifan Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yunjie Song
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Xingxing Cheng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Xu Guo
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Shengran Yun
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yanan Lu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Mingjie Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Junya Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266200, China.
| |
Collapse
|
46
|
Stringham NT, Green M, Roche W, Prado-Cabrero A, Mulcahy R, Nolan J. Lutein, zeaxanthin, and meso-zeaxanthin supplementation attenuates inflammatory cytokines and markers of oxidative cardiovascular processes in humans. Nutr Metab Cardiovasc Dis 2024; 34:1976-1983. [PMID: 38890092 DOI: 10.1016/j.numecd.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Systemic inflammation and oxidation are primary contributors to the development of atherosclerosis. Oxidation of low-density lipoprotein (LDL) particles within the vascular endothelium has been hypothesized to be an initial step in the formation of atherosclerotic plaques, with inflammatory cytokines serving as the signaling mechanism for concomitant macrophage activation. Supplementation with the antioxidative macular xanthophylls (lutein [L], zeaxanthin [Z], and meso-zeaxanthin [MZ]) has been shown to aid in the reduction of inflammatory physiologic responses; therefore, we hypothesized that in our study population, supplementation with these xanthophylls would facilitate a systemic reduction in markers of inflammation and cardiovascular lipid oxidation. METHODS AND RESULTS In this double-blind placebo-controlled supplementation study, participants were randomly allocated to receive the active intervention containing L (10 mg) + MZ (10 mg) + Z (2 mg) or placebo (containing sunflower oil). Serum concentrations of carotenoids (assessed by HPLC), inflammatory cytokines (IL-6, IL-1β, TNF-α) and oxidized LDL (OxLDL; by solid-phase sandwich ELISA) were measured at baseline and at 6-months. Results showed that over the supplementation period, compared to placebo, the active group demonstrated statistically significant increases in serum concentrations of L, Z, & MZ (p < 0.05), reductions in inflammatory cytokines IL-1β (p < 0.001) and TNF-α (p = 0.003), as well as a corresponding reduction in serum OxLDL (p = 0.009). CONCLUSIONS Our data show that L, Z, & MZ supplementation results in decreased serum IL-1β, TNF-α, and OxLDL. This suggests that these carotenoids are acting systemically to attenuate oxidative lipid products and inflammation, thus reducing their contribution to atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Nicole T Stringham
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland; Northern Arizona University, Flagstaff, AZ, USA.
| | - Marina Green
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Warren Roche
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Alfonso Prado-Cabrero
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Riona Mulcahy
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - John Nolan
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| |
Collapse
|
47
|
Ryan A, Rahman S, Williams RM. Optical Aptamer-Based Cytokine Nanosensor Detects Macrophage Activation by Bacterial Toxins. ACS Sens 2024; 9:3697-3706. [PMID: 38934367 PMCID: PMC11287749 DOI: 10.1021/acssensors.4c00887] [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: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Overactive or dysregulated cytokine expression is a hallmark of many acute and chronic inflammatory diseases. This is true for acute or chronic infections, neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, cancer, and others. Cytokines such as interleukin-6 (IL-6) are known therapeutic targets and biomarkers for such inflammatory diseases. Platforms for cytokine detection are, therefore, desirable tools for both research and clinical applications. Single-walled carbon nanotubes (SWCNT) are versatile nanomaterials with near-infrared fluorescence that can serve as transducers for optical sensors. When functionalized with an analyte-specific recognition element, SWCNT emission may become sensitive and selective toward the desired target. SWCNT-aptamer sensors are easily assembled, inexpensive, and biocompatible. In this work, we introduced a nanosensor design based on SWCNT and a DNA aptamer specific to IL-6. We first evaluated several SWCNT-aptamer constructs based on this simple direct complexation method, wherein the aptamer both solubilizes the SWCNT and confers sensitivity to IL-6. The sensor limit of detection, 105 ng/mL, lies in the relevant range for pathological IL-6 levels. Upon investigation of sensor kinetics, we found rapid response within seconds of antigen addition which continued over the course of 3 h. We found that this sensor construct is stable and the aptamer is not displaced from the nanotube surface during IL-6 detection. Finally, we investigated the ability of this sensor construct to detect macrophage activation caused by bacterial lipopolysaccharides (LPS) in an in vitro model of disease, finding rapid and sensitive detection of macrophage-expressed IL-6. We are confident that further development of this sensor will have novel implications for diagnosis of acute and chronic inflammatory diseases, in addition to contributing to the understanding of the role of cytokines in these diseases.
Collapse
Affiliation(s)
- Amelia
K. Ryan
- Department
of Biomedical Engineering, The City College
of New York, New York, New York 10031, United States
| | - Syeda Rahman
- Department
of Biomedical Engineering, The City College
of New York, New York, New York 10031, United States
| | - Ryan M. Williams
- Department
of Biomedical Engineering, The City College
of New York, New York, New York 10031, United States
- PhD
Program in Chemistry, Graduate Center, City
University of New York, New York, New York 10016, United States
| |
Collapse
|
48
|
Rahman MF, Kurlovs AH, Vodnala M, Meibalan E, Means TK, Nouri N, de Rinaldis E, Savova V. Immune disease dialogue of chemokine-based cell communications as revealed by single-cell RNA sequencing meta-analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603936. [PMID: 39071425 PMCID: PMC11275869 DOI: 10.1101/2024.07.17.603936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Immune-mediated diseases are characterized by aberrant immune responses, posing significant challenges to global health. In both inflammatory and autoimmune diseases, dysregulated immune reactions mediated by tissue-residing immune and non-immune cells precipitate chronic inflammation and tissue damage that is amplified by peripheral immune cell extravasation into the tissue. Chemokine receptors are pivotal in orchestrating immune cell migration, yet deciphering the signaling code across cell types, diseases and tissues remains an open challenge. To delineate disease-specific cell-cell communications involved in immune cell migration, we conducted a meta-analysis of publicly available single-cell RNA sequencing (scRNA-seq) data across diverse immune diseases and tissues. Our comprehensive analysis spanned multiple immune disorders affecting major organs: atopic dermatitis and psoriasis (skin), chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis (lung), ulcerative colitis (colon), IgA nephropathy and lupus nephritis (kidney). By interrogating ligand-receptor (L-R) interactions, alterations in cell proportions, and differential gene expression, we unveiled intricate disease-specific and common immune cell chemoattraction and extravasation patterns. Our findings delineate disease-specific L-R networks and shed light on shared immune responses across tissues and diseases. Insights gleaned from this analysis hold promise for the development of targeted therapeutics aimed at modulating immune cell migration to mitigate inflammation and tissue damage. This nuanced understanding of immune cell dynamics at the single-cell resolution opens avenues for precision medicine in immune disease management.
Collapse
Affiliation(s)
- Mouly F. Rahman
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Andre H. Kurlovs
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Munender Vodnala
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Elamaran Meibalan
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Terry K. Means
- Immunology & Inflammation Research Therapeutic Area, Sanofi US, Cambridge, MA 02141, United States
| | - Nima Nouri
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Emanuele de Rinaldis
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| | - Virginia Savova
- Precision Medicine and Computational Biology, Sanofi US, Cambridge, MA 02141, United States
| |
Collapse
|
49
|
Li F, Tan Z, Chen H, Gao Y, Xia J, Huang T, Liang L, Zhang J, Zhang X, Shi X, Chen Q, Shu Q, Yu L. Integrative analysis of bulk and single-cell RNA sequencing reveals the gene expression profile and the critical signaling pathways of type II CPAM. Cell Biosci 2024; 14:94. [PMID: 39026356 PMCID: PMC11264590 DOI: 10.1186/s13578-024-01276-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUD Type II congenital pulmonary airway malformation (CPAM) is a rare pulmonary microcystic developmental malformation. Surgical excision is the primary treatment for CPAM, although maternal steroids and betamethasone have proven effective in reducing microcystic CPAM. Disturbed intercellular communication may contribute to the development of CPAM. This study aims to investigate the expression profile and analyze intercellular communication networks to identify genes potentially associated with type II CPAM pathogenesis and therapeutic targets. METHODS RNA sequencing (RNA-seq) was performed on samples extracted from both the cystic area and the adjacent normal tissue post-surgery in CPAM patients. Iterative weighted gene correlation network analysis (iWGCNA) was used to identify genes specifically expressed in type II CPAM. Single-cell RNA-seq (scRNA-seq) was integrated to unveil the heterogeneity in cell populations and analyze the communication and interaction within epithelial cell sub-populations. RESULTS A total of 2,618 differentially expressed genes were identified, primarily enriched in cilium-related biological process and inflammatory response process. Key genes such as EDN1, GPR17, FPR2, and CHRM1, involved in the G protein-coupled receptor (GPCR) signaling pathway and playing roles in cell differentiation, apoptosis, calcium homeostasis, and the immune response, were highlighted based on the protein-protein interaction network. Type II CPAM-associated modules, including ciliary function-related genes, were identified using iWGCNA. By integrating scRNA-seq data, AGR3 (related to calcium homeostasis) and SLC11A1 (immune related) were identified as the only two differently expressed genes in epithelial cells of CPAM. Cell communication analysis revealed that alveolar type 1 (AT1) and alveolar type 2 (AT2) cells were the predominant communication cells for outgoing and incoming signals in epithelial cells. The ligands and receptors between epithelial cell subtypes included COLLAGEN genes enriched in PI3K-AKT singaling and involved in epithelial to mesenchymal transition. CONCLUSIONS In summary, by integrating bulk RNA-seq data of type II CPAM with scRNA-seq data, the gene expression profile and critical signaling pathways such as GPCR signaling and PI3K-AKT signaling pathways were revealed. Abnormally expressed genes in these pathways may disrupt epithelial-mesenchymal transition and contribute to the development of CPAM. Given the effectiveness of prenatal treatments of microcystic CPAM using maternal steroids and maternal betamethasone administration, targeting the genes and signaling pathways involved in the development of CPAM presents a promising therapeutic strategy.
Collapse
Affiliation(s)
- Fengxia Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Zheng Tan
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Hongyu Chen
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Yue Gao
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jie Xia
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Ting Huang
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Liang Liang
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jian Zhang
- Department of Thoracic Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xianghong Zhang
- Department of Cardiac Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xucong Shi
- Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Qiang Chen
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Jiangxi, China.
| | - Qiang Shu
- Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Lan Yu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| |
Collapse
|
50
|
Naraki K, Ghasemzadeh Rahbardar M, Razavi BM, Aminifar T, Khajavi Rad A, Amoueian S, Hosseinzadeh H. The power of trans-sodium crocetinate: exploring its renoprotective effects in a rat model of colistin-induced nephrotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03259-5. [PMID: 38995374 DOI: 10.1007/s00210-024-03259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024]
Abstract
Colistin, a multidrug-resistant gram-negative bacterial infection medication, has been associated with renal impairment and failure. Trans-sodium crocetinate (TSC), a saffron-derived chemical recognized for its antioxidant and nephroprotective properties, was studied in this study to determine its potential to alleviate the nephrotoxic effects of colistin. Forty-two male Wistar rats were randomly classified into seven groups (n = 6): (1) control (normal saline, 12 days, i.p.), (2) colistin (22 mg/kg, 7 days, i.p.), (3-5) colistin + TSC (25, 50, and 100 mg/kg, 12 days, i.p., starting from 5 days before colistin), (6) TSC (100 mg/kg, 12 days, i.p.), (7) colistin + vitamin E (100 IU/kg, 12 days, i.p). On day 13, the rats were euthanized and the serum content of creatinine, BUN, Na+, and K+, as well as oxidative stress (GSH, MDA, SOD, CAT), inflammatory (IL-1β), apoptotic (Bax, Bcl-2, caspase-3, 8, 9), and autophagy (Beclin-1, LC3) markers, NGAL, and histopathological changes in the kidney were measured. Colistin significantly increased serum creatinine, BUN, MDA, IL-1β, caspase-3,8,9, Bax, Beclin-1, LC3, and NGAL levels in kidney tissue. It also caused inflammation, focal necrosis of tubular epithelial cells, protein cast, and acute tubular necrosis. Furthermore, colistin decreased SOD, CAT, GSH, and Bcl-2 levels. TSC and vitamin E administration along with colistin restored most of the alterations induced by colistin. Overall, it could be concluded that colistin induces oxidative stress, inflammation, autophagy, and apoptosis, which can cause kidney injury. However, TSC can also be used as a therapeutic agent to reduce injuries caused by colistin.
Collapse
Affiliation(s)
- Karim Naraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tahereh Aminifar
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Khajavi Rad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sakineh Amoueian
- Pathology Department, Emam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|