1
|
Israeli Dangoor S, Khoury R, Salomon K, Pozzi S, Shahar S, Miari A, Leichtmann-Bardoogo Y, Bar-Hai N, Frommer N, Yeini E, Winkler T, Balint Lahat N, Kamer I, Hadad O, Laue K, Brem H, Hyde TM, Bar J, Barshack I, Ben-David U, Ishay-Ronen D, Maoz BM, Satchi-Fainaro R. CCL2 blockade combined with PD-1/P-selectin immunomodulators impedes breast cancer brain metastasis. Brain 2025; 148:1740-1756. [PMID: 39450648 PMCID: PMC12073999 DOI: 10.1093/brain/awae347] [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: 03/30/2024] [Revised: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Over the last two decades, the diagnosis and treatment of breast cancer patients have improved considerably. However, brain metastases remain a major clinical challenge and a leading cause of mortality. Thus, a better understanding of the pathways involved in the metastatic cascade is essential. To this end, we have investigated the reciprocal effects of astrocytes and breast cancer cells, employing traditional 2D cell culture and our unique 3D multicellular tumouroid models. Our findings revealed that astrocytes enhance the proliferation, migration and invasion of breast cancer cells, suggesting a supportive role for astrocytes in breast cancer outgrowth to the brain. Elucidating the key players in astrocyte-breast cancer cells crosstalk, we found that CCL2 is highly expressed in breast cancer brain metastases tissue sections from both patients and mice. Our in vitro and in vivo models further confirmed that CCL2 has a functional role in brain metastasis. Given their aggressive nature, we sought additional immune checkpoints for rationale combination therapy. Among the promising candidates were the adhesion molecule P-selectin, which we have recently shown to play a key role in the crosstalk with microglia cells and the co-inhibitory receptor PD-1, the main target of currently approved immunotherapies. Finally, combining CCL2 inhibition with immunomodulators targeting either PD-1/PD-L1 or P-selectin/P-Selectin Ligand-1 axes in our human 3D tumouroid models and in vivo presented more favourable outcomes than each monotherapy. Taken together, we propose that CCL2-CCR2/CCR4 is a key pathway promoting breast cancer brain metastases and a promising target for an immunotherapeutic combination approach.
Collapse
Affiliation(s)
- Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Koren Salomon
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shir Shahar
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adan Miari
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Neta Bar-Hai
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Neta Frommer
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tom Winkler
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Nora Balint Lahat
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
| | - Iris Kamer
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Ori Hadad
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Kathrin Laue
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21218, USA
- Department of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jair Bar
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Ishay-Ronen
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
2
|
Yuan YT, Guo TC, Wu CY, Yeh YQ, Wu CT, Hsu SY, Weng ZW, Ho MR, Hsu STD, Chen YC, Chang CR, Wu KP, Jeng US, Sue SC. Filamentous chemokine CCL5 structure and the functional aspects. Sci Rep 2025; 15:13552. [PMID: 40253490 PMCID: PMC12009402 DOI: 10.1038/s41598-025-98114-9] [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: 12/14/2024] [Accepted: 04/09/2025] [Indexed: 04/21/2025] Open
Abstract
Human inflammation-related CC chemokine ligand 5 (hCCL5) has significant self-assembly property under physiological conditions. The mechanism and function of hCCL5 oligomerization remain unclear. Different intermolecular interactions, such as E66-K25 or E66-R44/K45, have been reported to mediate hCCL5 oligomerization. This complexity makes structural determination difficult. Based on a K25S mutation to eliminate the E66-K25 interaction, we observed hCCL5 forming a helical-sharped filament in transmission electron microscopy (TEM). The filamentous polymerization is a dominant process when the concentration reaches ~ 100 nM and the filaments further form a higher-order assembly when concentration increases. In this large filament, a combination of X-ray solution scattering and cryo-EM analysis determined the structure; NMR further confirmed the filament packing, in which the interactions of residues R44 and K45 are critically involved. The sequence 43TRKNR47 was found to be essential for CCL5 trafficking inside the cells and for glycosaminoglycan binding outside the cells. The functional aspects of the chemokine filament are discussed.
Collapse
Affiliation(s)
- Yi-Ting Yuan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Tzu-Ching Guo
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chu-Ya Wu
- Instrumentation Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Qi Yeh
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Ching-Tse Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Yao Hsu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Zi-Wen Weng
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Meng-Ru Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, Higashihiroshima, Japan
| | - Yi-Chen Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chuang-Rung Chang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Kuen-Phon Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| | - U-Ser Jeng
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
- College of Semiconductor Research, National Tsing Hua University, Hsinchu, Taiwan.
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan.
| | - Shih-Che Sue
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
3
|
Kuraeiad S, Kotepui KU, Mahittikorn A, Anabire NG, Masangkay FR, Wilairatana P, Wangdi K, Kotepui M. A Systematic Review and Meta-Analysis of MIP-1α and MIP-1β Chemokines in Malaria in Relation to Disease Severity. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:676. [PMID: 40282967 PMCID: PMC12028554 DOI: 10.3390/medicina61040676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Macrophage inflammatory protein-1α (MIP-1α) and MIP-1β act as signaling molecules that recruit immune cells to sites of infection and inflammation. This study aimed to synthesize evidence on blood levels of MIP-1α and MIP-1β in Plasmodium-infected individuals and to determine whether these levels differ between severe and uncomplicated malaria cases. Materials and Methods: The study protocol was registered in PROSPERO (CRD42024595818). Comprehensive literature searches were conducted in six databases (EMBASE, MEDLINE, Ovid, Scopus, ProQuest, and PubMed) to identify studies reporting blood levels of MIP-1α and MIP-1β in Plasmodium infections and clinical malaria. A narrative synthesis was used to describe variations in MIP-1α and MIP-1β levels between malaria patients and controls and between severe and non-severe malaria cases. Meta-analysis was used to aggregate quantitative data utilizing a random-effects model. Results: A total of 1638 records were identified, with 20 studies meeting the inclusion criteria. Most studies reported significantly higher MIP-1α and MIP-1β levels in malaria patients compared to non-malarial controls. The meta-analysis showed a significant elevation in MIP-1α levels in malaria patients (n = 352) compared to uninfected individuals (n = 274) (p = 0.0112, random effects model, standardized mean difference [SMD]: 1.69, 95% confidence interval [CI]: 0.38 to 3.00, I2: 96.0%, five studies, 626 individuals). The meta-analysis showed no difference in MIP-1α levels between severe malaria cases (n = 203) and uncomplicated cases (n = 106) (p = 0.51, SMD: -0.48, 95% CI: -1.93 to 0.96, I2: 97.3%, three studies, 309 individuals). Conclusions: This study suggests that while MIP-1α and MIP-1β levels are elevated in malaria patients compared to uninfected individuals, these chemokines show a limited ability to differentiate between severe and uncomplicated malaria or predict severe outcomes. Further research is needed to clarify their role in malaria pathogenesis and explore potential clinical applications.
Collapse
Affiliation(s)
- Saruda Kuraeiad
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat 80160, Thailand;
- Center of Excellence in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | | | - Aongart Mahittikorn
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Nsoh Godwin Anabire
- Department of Biochemistry & Molecular Medicine, School of Medicine, University for Development Studies, Tamale P.O. Box TL 1882, Ghana
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell & Molecular Biology, University of Ghana, Accra P.O. Box LG 226, Ghana
| | | | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Kinley Wangdi
- HEAL Global Research Centre, Health Research Centre, Faculty of Health, University of Canberra, Canberra, ACT 2617, Australia
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia
| | - Manas Kotepui
- Medical Technology, Faculty of Science, Nakhon Phanom University, Nakhon Phanom 48000, Thailand
| |
Collapse
|
4
|
Diao S, Li L, Zhang J, Ji M, Sun L, Shen W, Wu S, Chen Z, Huang C, Li J. Macrophage-derived CCL1 targets CCR8 receptor in hepatic stellate cells to promote liver fibrosis through JAk/STAT pathway. Biochem Pharmacol 2025; 237:116884. [PMID: 40122149 DOI: 10.1016/j.bcp.2025.116884] [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: 06/05/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Liver fibrosis is caused by liver injury resulting from the wound healing response. According to recent research, the primary factor responsible for liver fibrosis is the activation of hepatic stellate cells (HSCs). C-C motif chemokine ligand 1 (CCL1) is one of several chemokine genes clustered on chromosome 17, which is involved in immune regulation and inflammatory processes. However, the role of CCL1 in liver fibrosis has not been reported. We found that CCL1 secreted by macrophages can target and activate the receptor protein C-C motif chemokine receptor 8 (CCR8) of HSCs, accelerating liver fibrosis progression by activating the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway. This suggested that the CCL1-mediated regulation of CCR8 is an important event in liver fibrosis progression. In conclusion, this study identified a novel signalling axis, the CCL1/CCR8/JAK/STAT pathway, which regulates the activation and apoptosis of HSCs, thus providing a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Shaoxi Diao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Liangyun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Jintong Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Minglu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Lijiao Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Wenwen Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Zixiang Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China.
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China.
| |
Collapse
|
5
|
Van Damme J, Struyf S, Proost P, Opdenakker G, Gouwy M. Functional Interactions Between Recombinant Serum Amyloid A1 (SAA1) and Chemokines in Leukocyte Recruitment. Int J Mol Sci 2025; 26:2258. [PMID: 40076881 PMCID: PMC11900440 DOI: 10.3390/ijms26052258] [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: 12/31/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The acute phase response is a hallmark of all inflammatory reactions and acute phase reactants, such as C-reactive protein (CRP) and serum amyloid A (SAA) proteins, are among the most useful plasma and serum markers of inflammation in clinical medicine. Although it is well established that inflammatory cytokines, mainly interleukin-1 (IL-1), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) induce SAA in the liver, the biological functions of elicited SAA remain an enigma. By the classical multi-step protein purification studies of chemotactic factors present in plasma or serum, we discovered novel chemokines and SAA1 fragments, which are induced during inflammatory reactions. In contrast to earlier literature, pure SAA1 fails to induce chemokines, an ascribed function that most probably originates from contaminating lipopolysaccharide (LPS). However, intact SAA1 and fragments thereof synergize with CXC and CC chemokines to enhance chemotaxis. Natural SAA1 fragments are generated by inflammatory proteinases such as matrix metalloproteinase-9 (MMP-9). They mediate synergy with chemokines by the interaction with cognate G protein-coupled receptors (GPCRs), formyl peptide receptor 2 (FPR2) and (CC and CXC) chemokine receptors. In conclusion, SAA1 enforces the action of many chemokines and assists in local leukocyte recruitment, in particular, when the concentrations of specifically-induced chemokines are still low.
Collapse
Affiliation(s)
| | | | | | | | - Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium; (J.V.D.); (S.S.); (P.P.); (G.O.)
| |
Collapse
|
6
|
Opgenorth J, Goetz BM, Rodriguez-Jimenez S, Freestone AD, Combs GJ, Flemming TA, McGill JL, Gorden PJ, Tikofsky L, Baumgard LH. Comparing oral versus intravenous calcium administration on alleviating markers of production, metabolism, and inflammation during an intravenous lipopolysaccharide challenge in mid-lactation dairy cows. J Dairy Sci 2025; 108:2883-2896. [PMID: 39603495 DOI: 10.3168/jds.2024-24831] [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: 02/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Animals, including dairy cows, develop hypocalcemia during infection. Prior independent research suggests supplementing oral Ca, but not i.v. Ca, improves multiple health metrics after immune activation. Therefore, study objectives were to directly compare the effects of administering an oral Ca bolus versus i.v. Ca on mineral and energetic metabolism variables and inflammatory parameters following an i.v. LPS challenge. Mid-lactation cows (124 ± 43 DIM) were assigned to 1 of 4 treatments: (1) saline control (CON; 4 mL of saline; n = 4), (2) LPS control (CON-LPS; 0.375 µg/kg BW; n = 6), (3) LPS with oral Ca bolus (OCa-LPS; 0.375 µg/kg BW and a 192-g bolus of Bovikalc [Boehringer Ingelheim Animal Health USA Inc., Duluth, GA] containing 43 g of Ca [71% CaCl2 and 29% CaSO4] supplemented at -0.5 and 6 h relative to LPS administration; n = 8), and (4) LPS with i.v. Ca (IVCa-LPS; 0.375 µg/kg BW and 500 mL of Ca-gluconate, 23% [VetOne, Boise, ID]) supplemented at -0.5 and 6 h relative to LPS infusion; n = 8). During period (P) 1 (4 d), baseline data were obtained. At the initiation of P2 (5 d), LPS and Ca supplements were administered. As anticipated, CON-LPS became hypocalcemic, but OCa-LPS and IVCa-LPS had increased ionized Ca compared with CON-LPS cows (1.11 and 1.28 vs. 0.95 ± 0.02 mmol/L, respectively). Rectal temperature increased after LPS and was additionally elevated in IVCa-LPS from 3 to 4 h (38.9 and 39.8 ± 0.1°C in CON-LPS and IVCa-LPS, respectively). Administering LPS decreased DMI and milk yield relative to CON. Circulating glucose was decreased in OCa-LPS compared with CON-LPS and IVCa-LPS during the initial hyperglycemic phase at 1 h (75.1 vs. 94.9 and 95.7 ± 3.4 mg/dL, respectively, but all LPS infused cows regardless of treatment had similar glucose concentrations thereafter, which were decreased relative to baseline during the first 12 h. Blood urea nitrogen increased after LPS but this was attenuated in OCa-LPS compared with CON-LPS and IVCa-LPS cows (8.7 vs. 10.0 and 10.4 ± 0.3 mg/dL). Glucagon increased in OCa-LPS and IVCa-LPS compared with CON-LPS cows (459 and 472 vs. 335 ± 28 pg/mL, respectively), and insulin markedly increased over time regardless of LPS treatment. Lipopolysaccharide substantially increased serum amyloid A, LPS-binding protein (LBP), and haptoglobin in all treatments, but OCa-LPS tended to have increased LBP concentrations relative to IVCa-LPS (10.7 vs. 8.6 ± 0.7 µg/mL, respectively). Several cytokines increased after LPS administration, but most temporal cytokine profiles did not differ by treatment. In summary, LPS administration intensely activated the immune system and both Ca delivery routes successfully ameliorated the hypocalcemia. The i.v. and oral Ca treatments had differential effects on multiple metabolism variables and appeared to mildly influence production responses to LPS.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - G J Combs
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - T A Flemming
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - J L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011
| | - P J Gorden
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA 50011
| | - L Tikofsky
- Boehringer Ingelheim Animal Health USA Inc., Duluth, GA 30096
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
7
|
Wu W, Li Q. Mechanisms of hydrocephalus after intraventricular haemorrhage: a review. Childs Nerv Syst 2024; 41:49. [PMID: 39674974 DOI: 10.1007/s00381-024-06711-2] [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: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Intraventricular haemorrhage (IVH) is bleeding within the ventricular system, which in adults is usually mainly secondary to cerebral haemorrhage and subarachnoid haemorrhage. Hydrocephalus is one of the most common complications of intraventricular haemorrhage, which is characterised by an increase in intracranial pressure due to an increased accumulation of cerebrospinal fluid within the ventricular system, and is closely related to the patient's prognosis. Surgical methods such as shunt surgery have been used to treat secondary hydrocephalus in recent years and have been effective in improving the survival and prognosis of patients with hydrocephalus. However, complications such as shunt blockage and intracranial infection are often faced after surgery. Moreover, little is known about the mechanism of hydrocephalus secondary to intraventricular haemorrhage. This review discusses the mechanisms regarding the occurrence of secondary hydrocephalus after intraventricular haemorrhage in adults in terms of blood clot obstruction, altered cerebrospinal fluid dynamics, inflammation, and blood composition.
Collapse
Affiliation(s)
- Wenchao Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 157, Health Care RoadHeilongjiang Province, Harbin City, Harbin, China
| | - Qingsong Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 157, Health Care RoadHeilongjiang Province, Harbin City, Harbin, China.
| |
Collapse
|
8
|
Pezeshkian F, Shahriarirad R, Mahram H. An overview of the role of chemokine CX3CL1 (Fractalkine) and CX3C chemokine receptor 1 in systemic sclerosis. Immun Inflamm Dis 2024; 12:e70034. [PMID: 39392260 PMCID: PMC11467895 DOI: 10.1002/iid3.70034] [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: 02/02/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) is a complex autoimmune disease characterized by fibrosis, vascular damage, and immune dysregulation. Fractalkine or chemokine (C-X3-C motif) ligand 1 (CX3CL1), a chemokine and adhesion molecule, along with its receptor CX3CR1, have been implicated in the inflammatory processes of SSc. CX3CL1 functions as both a chemoattractant and an adhesion molecule, guiding immune cell trafficking. This systematic review examines the role of CX3CL1 and its receptor CX3CR1 in the pathogenesis of SSc, with a focus on pulmonary and vascular complications. METHODS A systematic literature search was conducted across databases including PubMed, Scopus, and Web of Science from inception to November 2020. The search focused on studies investigating the CX3CL1/CX3CR1 axis in the context of SSc. RESULTS The review identified elevated CX3CL1 expression in SSc patients, particularly in the skin and lungs, where CX3CR1 is expressed on infiltrating immune cells. Higher levels of CX3CL1 were correlated with the severity of interstitial lung disease in SSc patients, indicating a potential predictive marker for disease progression. CX3CR1-positive monocytes and NK cells were recruited to inflamed tissues, contributing to fibrosis and tissue damage. Animal studies showed that inhibition of the CX3CL1/CX3CR1 axis reduced fibrosis and improved vascular function. CONCLUSION The CX3CL1/CX3CR1 axis plays a key role in immune cell recruitment and fibrosis in SSc. Elevated CX3CL1 levels are associated with lung and vascular complications, making it a potential biomarker for disease progression and a promising therapeutic target.
Collapse
Affiliation(s)
| | - Reza Shahriarirad
- Thoracic and Vascular Surgery Research CenterShiraz University of Medical SciencesShirazIran
| | | |
Collapse
|
9
|
Kondo S, Shiga S, Sakurai T. Elastin microfibril interface-located protein 1 in fibroblasts is regulated by amphiregulin and interleukin-1α produced by keratinocytes. Int J Cosmet Sci 2024; 46:680-690. [PMID: 38356201 DOI: 10.1111/ics.12947] [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: 09/29/2023] [Revised: 12/07/2023] [Accepted: 01/07/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVE The structure of elastic fibres changes with ageing. Elastin microfibril interface-located protein 1 (EMILIN-1) is known to contribute to structural changes in elastic fibres. EMILIN-1 is one of the components of elastic fibres and also colocalizes with oxytalan fibres near the epidermis. Therefore, EMILIN-1 may be affected by epidermal-dermal interactions. The purpose of this study is to identify the key factors involved in epidermal-dermal interactions during the structural degeneration of elastic fibres. METHODS Keratinocytes and fibroblasts were co-cultured, and changes in elastic fibre-related proteins were evaluated. Additionally, cytokine arrays were used to identify the factors involved in epidermal-dermal interactions. RESULTS EMILIN-1 expression in fibroblasts was increased in the presence of keratinocytes, and its expression decreased when keratinocytes were stressed. Amphiregulin (AREG) and interleukin-1α (IL-1α) were identified as the keratinocyte-derived cytokines that influence the production of EMILIN-1, which is secreted by the fibroblasts. EMILIN-1 expression was promoted by AREG and decreased by IL-1α via an increase in cathepsin K (a catabolic enzyme). AREG and IL-1α were associated with changes in EMILIN-1 levels in fibroblasts. CONCLUSION The findings suggest that the suppression of IL-1α expression and promotion of AREG expression in the epidermis could be a new approach that prevents the wrinkles and sagging caused by the structural changes in elastic fibres.
Collapse
Affiliation(s)
- Shinya Kondo
- FANCL Research Institute, FANCL Corporation, Kanagawa, Japan
| | - Soichiro Shiga
- FANCL Research Institute, FANCL Corporation, Kanagawa, Japan
| | | |
Collapse
|
10
|
Kumar S, Mohan V, Kant Singh R, Kumar Gautam P, Kumar S, Shukla A, Kumar Patel A, Yadav L, Acharya A. Tumor-derived Hsp70-CD14 interaction enhances the antitumor potential of cytotoxic T cells by activating tumor-associated macrophages to express CC chemokines and CD40 costimulatory molecules. Int Immunopharmacol 2024; 138:112584. [PMID: 38944948 DOI: 10.1016/j.intimp.2024.112584] [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: 11/07/2023] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Heat shock proteins are a widely distributed group of proteins. It is constitutively expressed in almost all organisms and shows little variation throughout evolution. Previously, HSPs, particularly Hsp70, were recognized as molecular chaperones that aid in the proper three-dimensional folding of newly synthesized polypeptides in cells. Recently, researchers have focused on the potential induction of immune cells, including macrophages, antigen-specific CD8+ cytotoxic T cells, and PBMCs. It induces the expression of CC chemokines such as MIP-1α and RANTES, which are responsible for the chemotactic movement and migration of immune cells at the site of infection to neutralize foreign particles in vivo and in vitro in several cell lines but their effect on tumor-associated macrophages is still not known. These cytokines are also known to influence the movement of several immune cells, including CD8+ cytotoxic T cells, toward inflammatory sites. Therefore, the effect of tumor-derived autologous Hsp70 on the expression of MIP-lα and RANTES in tumor-associated macrophages (TAMs) was investigated. Our results indicated that Hsp70 treatment-induced MIP-lα and RANTES expression was significantly greater in TAMs than in NMOs. According to the literature, the CC chemokine shares the same receptor, CCR5, as HIV does for their action, and therefore could provide better completion to the virus for ligand binding. Furthermore, Hsp70-preactivated TAMs induced increased IL-2 and IFN-γ expression in T cells during coculture for 48 h and upregulated the antitumor immune response of the host. Therefore, the outcome of our study could be useful for developing a better approach to restricting the growth and progression of tumors.
Collapse
Affiliation(s)
- Sanjay Kumar
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Vijay Mohan
- School of Biological and Life Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishi Kant Singh
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | - Pramod Kumar Gautam
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sandeep Kumar
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Alok Shukla
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Anand Kumar Patel
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Lokesh Yadav
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Arbind Acharya
- Centre of Advanced Study, Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
11
|
Yuan Y. Imbalance of dendritic cell function in pulmonary fibrosis. Cytokine 2024; 181:156687. [PMID: 38963940 DOI: 10.1016/j.cyto.2024.156687] [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: 03/23/2024] [Revised: 05/08/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Pulmonary fibrosis (PF) is a chronic, irreversible interstitial lung disease. The pathogenesis of PF remains unclear, and there are currently no effective treatments or drugs that can completely cure PF. The primary cause of PF is an imbalance of inflammatory response and inappropriate repair following lung injury. Dendritic cells (DCs), as one of the immune cells in the body, play an important role in regulating immune response, immune tolerance, and promoting tissue repair following lung injury. However, the role of DCs in the PF process is ambiguous or even contradictory in the existing literature. On the one hand, DCs can secrete transforming growth factor β(TGF-β), stimulate Th17 cell differentiation, stimulate fibroblast proliferation, and promote the generation of inflammatory factors interleukin-6(IL-6) and tumor necrosis factor-α(TNF-α), thereby promoting PF. On the other hand, DCs suppress PF through mechanisms including the secretion of IL-10 to inhibit effector T cell activity in the lungs and promote the function of regulatory T cells (Tregs), as well as by expressing matrix metalloproteinases (MMPs) which facilitate the degradation of the extracellular matrix (ECM). This article will infer possible reasons for the different roles of DCs in PF and analyze possible reasons for the functional imbalance of DCs in pulmonary fibrosis from the complexity and changes of the pulmonary microenvironment, autophagy defects of DCs, and changes in the pulmonary physical environment.
Collapse
Affiliation(s)
- Yuan Yuan
- Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China.
| |
Collapse
|
12
|
Opgenorth J, Mayorga EJ, Abeyta MA, Goetz BM, Rodriguez-Jimenez S, Freestone AD, McGill JL, Baumgard LH. Intravenous lipopolysaccharide challenge in early- versus mid-lactation dairy cattle. I: The immune and inflammatory responses. J Dairy Sci 2024; 107:6225-6239. [PMID: 38428491 DOI: 10.3168/jds.2023-24350] [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: 10/25/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
Cows in early lactation (EL) are purportedly immune suppressed, which renders them more susceptible to disease. Thus, the study objective was to compare key biomarkers of immune activation from i.v. LPS between EL and mid-lactation (ML) cows. Multiparous EL (20 ± 2 DIM; n = 11) and ML (131 ± 31 DIM; n = 12) cows were enrolled in a 2 × 2 factorial design and assigned to 1 of 2 treatments by lactation stage (LS): (1) EL (EL-LPS; n = 6) or ML (ML-LPS; n = 6) cows administered a single LPS bolus from Escherichia coli O55:B5 (0.09 µg/kg of BW), or (2) pair-fed (PF) EL (EL-PF; n = 5) or ML (ML-PF; n = 6) cows administered i.v. saline. After LPS administration, cows were intensely evaluated for 3 d to analyze their response and recovery to LPS. Rectal temperature increased in LPS relative to PF cows (1.1°C in the first 9 h), and the response was more severe in EL-LPS relative to ML-LPS cows (2.3 vs. 1.3°C increase at 4 h post-LPS; respectively). Respiration rate increased only in EL-LPS cows (47% relative to ML-LPS in the first hour post-LPS). Circulating tumor necrosis factor-α, IL-6, monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1α, MIP-1β, and IFN-γ-inducible protein-10 increased within the first 6 h after LPS and these changes were exacerbated in EL-LPS relative to ML-LPS cows (6.3-fold, 4.8-fold, 57%, 93%, 10%, and 61%, respectively). All cows administered LPS had decreased circulating iCa relative to PF cows (34% at the 6 h nadir), but the hypocalcemia was more severe in EL-LPS than ML-LPS cows (14% at 6 h nadir). In response to LPS, neutrophils decreased regardless of LS, then increased into neutrophilia by 24 h in all LPS relative to PF cows (2-fold); however, the neutrophilic phase was augmented in EL- compared with ML-LPS cows (63% from 24 to 72 h). Lymphocytes and monocytes rapidly decreased then gradually returned to baseline in LPS cows regardless of LS; however, monocytes were increased (57%) at 72 h in EL-LPS relative to ML-LPS cows. Platelets were reduced (46%) in LPS relative to PF cows throughout the 3-d following LPS, and from 24 to 48 h, platelets were further decreased (41%) in EL-LPS compared with ML-LPS. During the 3-d following LPS, serum amyloid A (SAA), LPS-binding protein (LBP), and haptoglobin (Hp) increased in LPS compared with PF groups (9-fold, 72%, and 153-fold, respectively), and the LBP and Hp responses were more exaggerated in EL-LPS than ML-LPS cows (85 and 79%, respectively) whereas the SAA response did not differ by LS. Thus, our data indicates that EL immune function does not appear "suppressed," and in fact many aspects of the immune response are seemingly functionally robust.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - E J Mayorga
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - J L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
13
|
Yuan F, Zhang R, Li J, Lei Q, Wang S, Jiang F, Guo Y, Xiang M. CCR5-overexpressing mesenchymal stem cells protect against experimental autoimmune uveitis: insights from single-cell transcriptome analysis. J Neuroinflammation 2024; 21:136. [PMID: 38802924 PMCID: PMC11131209 DOI: 10.1186/s12974-024-03134-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
Autoimmune uveitis is a leading cause of severe vision loss, and animal models provide unique opportunities for studying its pathogenesis and therapeutic strategies. Here we employ scRNA-seq, RNA-seq and various molecular and cellular approaches to characterize mouse models of classical experimental autoimmune uveitis (EAU), revealing that EAU causes broad retinal neuron degeneration and marker downregulation, and that Müller glia may act as antigen-presenting cells. Moreover, EAU immune response is primarily driven by Th1 cells, and results in dramatic upregulation of CC chemokines, especially CCL5, in the EAU retina. Accordingly, overexpression of CCR5, a CCL5 receptor, in mesenchymal stem cells (MSCs) enhances their homing capacity and improves their immunomodulatory outcomes in preventing EAU, by reducing infiltrating T cells and activated microglia and suppressing Nlrp3 inflammasome activation. Taken together, our data not only provide valuable insights into the molecular characteristics of EAU but also open an avenue for innovative MSC-based therapy.
Collapse
Affiliation(s)
- Fa Yuan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Rong Zhang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jiani Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Qiannan Lei
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Shuyi Wang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Fanying Jiang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yanan Guo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
14
|
Liu Z, Zhao C, Yu H, Zhang R, Xue X, Jiang Z, Ge Z, Xu Y, Zhang W, Lin L, Chen Z. MCP-3 as a prognostic biomarker for severe fever with thrombocytopenia syndrome: a longitudinal cytokine profile study. Front Immunol 2024; 15:1379114. [PMID: 38812521 PMCID: PMC11134196 DOI: 10.3389/fimmu.2024.1379114] [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: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction Severe fever with thrombocytopenia syndrome (SFTS) is characterized by a high mortality rate and is associated with immune dysregulation. Cytokine storms may play an important role in adverse disease regression, this study aimed to assess the validity of MCP-3 in predicting adverse outcomes in SFTS patients and to investigate the longitudinal cytokine profile in SFTS patients. Methods The prospective study was conducted at Yantai Qishan Hospital from May to November 2022. We collected clinical data and serial blood samples during hospitalization, patients with SFTS were divided into survival and non-survival groups based on the clinical prognosis. Results The levels of serum 48 cytokines were measured using Luminex assays. Compared to healthy controls, SFTS patients exhibited higher levels of most cytokines. The non-survival group had significantly higher levels of 32 cytokines compared to the survival group. Among these cytokines, MCP-3 was ranked as the most significant variable by the random forest (RF) model in predicting the poor prognosis of SFTS patients. Additionally, we validated the predictive effects of MCP-3 through receiver operating characteristic (ROC) curve analysis with an AUC of 0.882 (95% CI, 0.787-0.978, P <0.001), and the clinical applicability of MCP-3 was assessed favorably based on decision curve analysis (DCA). The Spearman correlation analysis indicated that the level of MCP-3 was positively correlated with ALT, AST, LDH, α-HBDH, APTT, D-dimer, and viral load (P<0.01). Discussion For the first time, our study identified and validated that MCP-3 could serve as a meaningful biomarker for predicting the fatal outcome of SFTS patients. The longitudinal cytokine profile analyzed that abnormally increased cytokines were associated with the poor prognosis of SFTS patients. Our study provides new insights into exploring the pathogenesis of cytokines with organ damage and leading to adverse effects.
Collapse
Affiliation(s)
- Zishuai Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chenxi Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hong Yu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Rongling Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Xue
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhouling Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziruo Ge
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanli Xu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Wei Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Zhihai Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Kłósek M, Kurek-Górecka A, Balwierz R, Krawczyk-Łebek A, Kostrzewa-Susłow E, Bronikowska J, Jaworska D, Czuba ZP. The Effect of Methyl-Derivatives of Flavanone on MCP-1, MIP-1β, RANTES, and Eotaxin Release by Activated RAW264.7 Macrophages. Molecules 2024; 29:2239. [PMID: 38792101 PMCID: PMC11123782 DOI: 10.3390/molecules29102239] [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: 03/11/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Chemokines, also known as chemotactic cytokines, stimulate the migration of immune cells. These molecules play a key role in the pathogenesis of inflammation leading to atherosclerosis, neurodegenerative disorders, rheumatoid arthritis, insulin-resistant diabetes, and cancer. Moreover, they take part in inflammatory bowel disease (IBD). The main objective of our research was to determine the activity of methyl-derivatives of flavanone, namely, 2'-methylflavanone (5B), 3'-methylflavanone (6B), 4'-methylflavanone (7B), and 6-methylflavanone (8B), on the releasing of selected cytokines by RAW264.7 macrophages activated by LPS. We determined the concentration of chemokines belonging to the CC chemokine family, namely, MCP-1, MIP-1β, RANTES, and eotaxin, using the Bio-Plex Magnetic Luminex Assay and the Bio-PlexTM 200 System. Among the tested compounds, only 5B and 6B had the strongest effect on inhibiting the examined chemokines' release by macrophages. Therefore, 5B and 6B appear to be potentially useful in the prevention of diseases associated with the inflammatory process.
Collapse
Affiliation(s)
- Małgorzata Kłósek
- Department of Microbiology and Immunology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland; (J.B.); (D.J.); (Z.P.C.)
| | - Anna Kurek-Górecka
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, St Kasztanowa 3, 41-200 Sosnowiec, Poland
| | - Radosław Balwierz
- Institute of Chemistry, University of Opole, Oleska 48, 45-052 Opole, Poland;
| | - Agnieszka Krawczyk-Łebek
- Department of Food Chemistry and Biocatalysis, University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland; (A.K.-Ł.); (E.K.-S.)
| | - Edyta Kostrzewa-Susłow
- Department of Food Chemistry and Biocatalysis, University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland; (A.K.-Ł.); (E.K.-S.)
| | - Joanna Bronikowska
- Department of Microbiology and Immunology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland; (J.B.); (D.J.); (Z.P.C.)
| | - Dagmara Jaworska
- Department of Microbiology and Immunology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland; (J.B.); (D.J.); (Z.P.C.)
| | - Zenon P. Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland; (J.B.); (D.J.); (Z.P.C.)
| |
Collapse
|
16
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [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: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
17
|
Chikata Y, Iwata H, Miyosawa K, Naito R, Koike T, Moriya S, Yasuda H, Funamizu T, Doi S, Endo H, Wada H, Ogita M, Dohi T, Kasai T, Isoda K, Okazaki S, Miyauchi K, Minamino T. Elevated levels of plasma inactive stromal cell derived factor-1α predict poor long-term outcomes in diabetic patients following percutaneous coronary intervention. Cardiovasc Diabetol 2024; 23:114. [PMID: 38555431 PMCID: PMC10981820 DOI: 10.1186/s12933-024-02197-z] [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: 10/26/2023] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Since the complication of diabetes mellitus (DM) is a risk for adverse cardiovascular outcomes in patients with coronary artery disease (CAD), appropriate risk estimation is needed in diabetic patients following percutaneous coronary intervention (PCI). However, there is no useful biomarker to predict outcomes in this population. Although stromal cell derived factor-1α (SDF-1α), a circulating chemokine, was shown to have cardioprotective roles, the prognostic impact of SDF-1α in diabetic patients with CAD is yet to be fully elucidated. Moreover, roles of SDF-1α isoforms in outcome prediction remain unclear. Therefore, this study aimed to assess the prognostic implication of three forms of SDF-1α including total, active, and inactive forms of SDF-1α in patients with DM and after PCI. METHODS This single-center retrospective analysis involved consecutive patients with diabetes who underwent PCI for the first time between 2008 and 2018 (n = 849). Primary and secondary outcome measures were all-cause death and the composite of cardiovascular death, non-fatal myocardial infarction, and ischemic stroke (3P-MACE), respectively. For determining plasma levels of SDF-1α, we measured not only total, but also the active type of SDF-1α by ELISA. Inactive isoform of the SDF-1α was calculated by subtracting the active isoform from total SDF-1α. RESULTS Unadjusted Kaplan-Meier analyses revealed increased risk of both all-cause death and 3P-MACE in patients with elevated levels of inactive SDF-1α. However, plasma levels of total and active SDF-1α were not associated with cumulative incidences of outcome measures. Multivariate Cox hazard analyses repeatedly indicated the 1 higher log-transformed inactive SDF-1α was significantly associated with increased risk of all-cause death (hazard ratio (HR): 2.64, 95% confidence interval (CI): 1.28-5.34, p = 0.008) and 3P-MACE (HR: 2.51, 95% CI: 1.12-5.46, p = 0.02). Moreover, the predictive performance of inactive SDF-1α was higher than that of total SDF-1α (C-statistics of inactive and total SDF-1α for all-cause death: 0.631 vs 0.554, for 3P-MACE: 0.623 vs 0.524, respectively). CONCLUSION The study results indicate that elevated levels of plasma inactive SDF-1α might be a useful indicator of poor long-term outcomes in diabetic patients following PCI. TRIAL REGISTRATION This study describes a retrospective analysis of a prospective registry database of patients who underwent PCI at Juntendo University Hospital, Tokyo, Japan (Juntendo Physicians' Alliance for Clinical Trials, J-PACT), which is publicly registered (University Medical Information Network Japan-Clinical Trials Registry, UMIN-CTR 000035587).
Collapse
Affiliation(s)
- Yuichi Chikata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hiroshi Iwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan.
| | | | - Ryo Naito
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takuma Koike
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Soshi Moriya
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hidetoshi Yasuda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takehiro Funamizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Shinichiro Doi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hirohisa Endo
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Hideki Wada
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Manabu Ogita
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Tomotaka Dohi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Shinya Okazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Katsumi Miyauchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| |
Collapse
|
18
|
Chapman-Lopez TJ, Funderburk LK, Heileson JL, Wilburn DT, Koutakis P, Gallucci AR, Forsse JS. Effects of L-Leucine Supplementation and Resistance Training on Adipokine Markers in Untrained Perimenopausal and Postmenopausal Women. J Strength Cond Res 2024; 38:526-532. [PMID: 38088883 DOI: 10.1519/jsc.0000000000004661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
ABSTRACT Chapman-Lopez, TJ, Funderburk, LK, Heileson, JL, Wilburn, DT, Koutakis, P, Gallucci, AR, and Forsse, JS. Effects of L-leucine supplementation and resistance training on adipokine markers in untrained perimenopausal and postmenopausal women. J Strength Cond Res 38(3): 526-532, 2024-This study examined the effects of supplementing 5 g of leucine compared with a placebo during a 10-week resistance training program on body composition parameters and adipokine concentrations in untrained, perimenopausal and postmenopausal women. Thirty-five women were randomly assigned to 2 groups-leucine (LEU, n = 17) and placebo (PLC, n = 18)-in a double-blind, placebo-controlled trial. Each group consumed the supplement or placebo every day and completed a resistance training program for 10 weeks. Using 3-day food records, a diet was assessed before the intervention and after its cessation. Body composition was assessed preintervention and postintervention using dual-energy x-ray absorptiometry. Moreover, the concentrations of adipokines, such as adiponectin, visfatin, leptin, and monocyte chemoattractant protein-1 (MCP-1), were assessed preintervention and postintervention. Both groups showed an increase in visceral adipose tissue (VAT) area ( p = 0.030) and fat-free mass (FFM; p = 0.023). There were significant group differences in concentrations of visfatin ( p = 0.020) and leptin ( p = 0.038) between the PLC and LEU groups. Visfatin displayed higher concentrations in the PLC group and leptin displayed higher concentrations in the LEU group. In addition, there were significant decreases in adiponectin concentrations for both groups (LEU: 652 ± 513 to 292 ± 447 pg·ml -1 ; PLC: 584 ± 572 to 245 ± 356 pg·ml -1 , p = 0.002) and MCP-1 only decreased in the PLC group (253 ± 119 to 206 ± 106 pg·ml -1 , p = 0.004). There were significant decreases in adiponectin concentrations in both groups and a decrease in MCP-1 concentrations in the PLC group. These decreases may be due to both adipokines possible relationship with VAT area. However, it is not known whether leucine has underlying properties that hinder changes in MCP-1 concentrations.
Collapse
Affiliation(s)
- Tomas J Chapman-Lopez
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
| | - LesLee K Funderburk
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
- Human Sciences and Design, Baylor University, Waco, Texas
| | - Jeffery L Heileson
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
- Nutrition Services Division, Walter Reed National Military Medical Center, Bethesda, Maryland; and
| | - Dylan T Wilburn
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
| | | | - Andrew R Gallucci
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
| | - Jeffrey S Forsse
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health Human Performance and Recreation, Baylor University, Waco, Texas
| |
Collapse
|
19
|
Ding C, Xiao T, Deng Y, Yang H, Xu B, Li J, Lv Z. The Teleost CXCL13-CXCR5 Axis Induces Inflammatory Cytokine Expression through the Akt-NF-κB, p38-AP-1, and p38-NF-κB Pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:317-334. [PMID: 38054894 DOI: 10.4049/jimmunol.2300237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
The ancestors of chemokines originate in the most primitive of vertebrates, which has recently attracted great interest in the immune functions and the underlying mechanisms of fish chemokines. In the current study, we identified an evolutionarily conserved chemokine, CiCXCL13, from a teleost fish, grass carp. CiCXCL13 was characterized by a typical SCY (small cytokine CXC) domain and four cysteine residues (C34, C36, C61, C77), with the first two cysteines separated by a random amino acid residue, although it shared 24.2-54.8% identity with the counterparts from other vertebrates. CiCXCL13 was an inducible chemokine, whose expression was significantly upregulated in the immune tissues of grass carps after grass carp reovirus infection. CiCXCL13 could bind to the membrane of grass carp head kidney leukocytes and promote cell migration, NO release, and the expression of >15 inflammatory cytokines, including IL-1β, TNF-α, IL-10 and TGF-β1, thus regulating the inflammatory response. Mechanistically, CiCXCL13 interacted with its evolutionarily conserved receptor CiCXCR5 and activated the Akt-NF-κB and p38-AP-1 pathways, as well as a previously unrevealed p38-NF-κB pathway, to efficiently induce inflammatory cytokine expression, which was distinct from that reported in mammals. Zebrafish CXCL13 induced inflammatory cytokine expression through Akt, p38, NF-κB, and AP-1 as CiCXCL13. Meanwhile, the CiCXCL13-CiCXCR5 axis-mediated inflammatory activity was negatively shaped by grass carp atypical chemokine receptor 2 (CiACKR2). The present study is, to our knowledge, the first to comprehensively define the immune function of CXCL13 in inflammatory regulation and the underlying mechanism in teleosts, and it provides a valuable perspective on the evolution and biology of fish chemokines.
Collapse
Affiliation(s)
- Chunhua Ding
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Yadong Deng
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Hong Yang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Baohong Xu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Junhua Li
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| |
Collapse
|
20
|
Al Hamwi G, Namasivayam V, Büschbell B, Gedschold R, Golz S, Müller CE. Proinflammatory chemokine CXCL14 activates MAS-related G protein-coupled receptor MRGPRX2 and its putative mouse ortholog MRGPRB2. Commun Biol 2024; 7:52. [PMID: 38184723 PMCID: PMC10771525 DOI: 10.1038/s42003-023-05739-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Patients with idiopathic pulmonary fibrosis show a strongly upregulated expression of chemokine CXCL14, whose target is still unknown. Screening of CXCL14 in a panel of human G protein-coupled receptors (GPCRs) revealed its potent and selective activation of the orphan MAS-related GPCR X2 (MRGPRX2). This receptor is expressed on mast cells and - like CXCL14 - upregulated in bronchial inflammation. CXCL14 induces robust activation of MRGPRX2 and its putative mouse ortholog MRGPRB2 in G protein-dependent and β-arrestin recruitment assays that is blocked by a selective MRGPRX2/B2 antagonist. Truncation combined with mutagenesis and computational studies identified the pharmacophoric sequence of CXCL14 and its presumed interaction with the receptor. Intriguingly, C-terminal domain sequences of CXCL14 consisting of 4 to 11 amino acids display similar or increased potency and efficacy compared to the full CXCL14 sequence (77 amino acids). These results provide a rational basis for the future development of potential idiopathic pulmonary fibrosis therapies.
Collapse
Affiliation(s)
- Ghazl Al Hamwi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Beatriz Büschbell
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Robin Gedschold
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Stefan Golz
- Lead Identification & Characterization, Pharma Research and Development Center, Bayer AG, Wuppertal, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
21
|
Yang M, Zhang C. The role of innate immunity in diabetic nephropathy and their therapeutic consequences. J Pharm Anal 2024; 14:39-51. [PMID: 38352948 PMCID: PMC10859537 DOI: 10.1016/j.jpha.2023.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/12/2023] [Accepted: 09/05/2023] [Indexed: 02/16/2024] Open
Abstract
Diabetic nephropathy (DN) is an enduring condition that leads to inflammation and affects a substantial number of individuals with diabetes worldwide. A gradual reduction in glomerular filtration and emergence of proteins in the urine are typical aspects of DN, ultimately resulting in renal failure. Mounting evidence suggests that immunological and inflammatory factors are crucial for the development of DN. Therefore, the activation of innate immunity by resident renal and immune cells is critical for initiating and perpetuating inflammation. Toll-like receptors (TLRs) are an important group of receptors that identify patterns and activate immune responses and inflammation. Meanwhile, inflammatory responses in the liver, pancreatic islets, and kidneys involve inflammasomes and chemokines that generate pro-inflammatory cytokines. Moreover, the activation of the complement cascade can be triggered by glycated proteins. This review highlights recent findings elucidating how the innate immune system contributes to tissue fibrosis and organ dysfunction, ultimately leading to renal failure. This review also discusses innovative approaches that can be utilized to modulate the innate immune responses in DN for therapeutic purposes.
Collapse
Affiliation(s)
- Min Yang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
22
|
Nowag B, Schäfer D, Hengl T, Corduff N, Goldie K. Biostimulating fillers and induction of inflammatory pathways: A preclinical investigation of macrophage response to calcium hydroxylapatite and poly-L lactic acid. J Cosmet Dermatol 2024; 23:99-106. [PMID: 37593832 DOI: 10.1111/jocd.15928] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Initial macrophage response to biostimulatory substances is key in determining the subsequent behavior of fibroblasts and the organization of newly synthesized collagen. Though histological studies suggest that calcium hydroxylapatite (CaHA) filler initiates a regenerative healing response with collagen and elastin deposition similar to natural, healthy tissue rather than an inflammatory response with fibrosis, the relative activity of macrophages stimulated by CaHA, as well as how this activity compares to that induced by other biostimulatory fillers, has not been explored. The aim of the study is to characterize the in vitro macrophage response to two biostimulory fillers, CaHA and PLLA (poly-L lactic acid), and to evaluate their inflammatory potential. METHODS Primary human macrophages were incubated with two dilutions (1:50 and 1:100) of commercially available CaHA or PLLA. After 24 h incubation, an inflammation array was used to screen for the expression of 40 cytokines, released by macrophages. ELISA was used to confirm array results. RESULTS Four cytokines were significantly upregulated in M1 macrophages incubated with PLLA compared to both unstimulated controls and CaHA: CCL1 (p < 0.001), TNFRII (p < 0.01), MIP-1α (p < 0.05), and IL-8 (p < 0.001). In M2 macrophages, MIP-1α (p < 0.01) and MIP-1β (p < 0.01) were significantly upregulated by PLLA compared to CaHA and unstimulated controls. CONCLUSION Together, these findings indicate that the CaHA mode of action is a non-inflammatory response while PLLA initiates expression of several cytokines known to play a role in inflammation. Our study supports the concept that these two "biostimulatory" fillers follow distinct pathways and should be considered individually with regard to mechanism of action.
Collapse
Affiliation(s)
| | | | - Thomas Hengl
- R&D, Merz Aesthetics GmbH, Frankfurt am Main, Germany
| | | | | |
Collapse
|
23
|
Guo X, Sun M, Yang P, Meng X, Liu R. Role of mast cells activation in the tumor immune microenvironment and immunotherapy of cancers. Eur J Pharmacol 2023; 960:176103. [PMID: 37852570 DOI: 10.1016/j.ejphar.2023.176103] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023]
Abstract
The mast cell is an important cellular component that plays a crucial role in the crosstalk between innate and adaptive immune responses within the tumor microenvironment (TME). Recently, numerous studies have indicated that mast cells related to tumors play a dual role in regulating cancers, with conflicting results seemingly determined by the degranulation medium. As such, mast cells are an ignored but very promising potential target for cancer immunotherapy based on their immunomodulatory function. In this review, we present a comprehensive overview of the roles and mechanisms of mast cells in diverse cancer types. Firstly, we evaluated the infiltration density and location of mast cells on tumor progression. Secondly, mast cells are activated by the TME and subsequently release a range of inflammatory mediators, cytokines, chemokines, and lipid products that modulate their pro-or anti-tumor functions. Thirdly, activated mast cells engage in intercellular communication with other immune or stromal cells to modulate the immune status or promote tumor development. Finally, we deliberated on the clinical significance of targeting mast cells as a therapeutic approach to restrict tumor initiation and progression. Overall, our review aims to provide insights for future research on the role of mast cells in tumors and their potential as therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China; Xiangnan University, Chenzhou, China
| | - Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Peiyan Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xingchen Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.
| |
Collapse
|
24
|
Wan S, Xie J, Liang Y, Yu X. Pathological roles of bone marrow adipocyte-derived monocyte chemotactic protein-1 in type 2 diabetic mice. Cell Death Discov 2023; 9:412. [PMID: 37957155 PMCID: PMC10643445 DOI: 10.1038/s41420-023-01708-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become a prevalent public health concern, with beta-cell dysfunction involved in its pathogenesis. Bone marrow adipose tissue (BMAT) increases in both the quantity and area in individuals with T2DM along with heightened monocyte chemotactic protein-1 (MCP-1) secretion. This study aims to investigate the influence and underlying mechanisms of MCP-1 originating from bone marrow adipocytes (BMAs) on systemic glucose homeostasis in T2DM. Initially, a substantial decrease in the proliferation and glucose-stimulated insulin secretion (GSIS) of islet cells was observed. Moreover, a comparative analysis between the control (Ctrl) group and db/db mice revealed significant alterations in the gene expression profiles of whole bone marrow cells, with a noteworthy upregulation of Mcp-1. And the primary enriched pathways included chemokine signaling pathway and AGE-RAGE signaling pathway in diabetic complications. In addition, the level of MCP-1 was distinctly elevated in BMA-derived conditional media (CM), leading to a substantial inhibition of proliferation, GSIS and the protein level of phosphorylated Akt (p-Akt) in Min6 cells. After blocking MCP-1 pathway, we observed a restoration of p-Akt and the proliferation of islet cells, resulting in a marked improvement in disordered glucose homeostasis. In summary, there is an accumulation of BMAs in T2DM, which secrete excessive MCP-1, exacerbating the abnormal accumulation of BMAs in the bone marrow cavity through paracrine signaling. The upregulated MCP-1, in turn, worsens glucose metabolism disorder by inhibiting the proliferation and insulin secretion of islet cells through an endocrine pathway. Inhibiting MCP-1 signaling can partially restore the proliferation and insulin secretion of islet cells, ultimately ameliorating glucose metabolism disorder. It's worth noting that to delve deeper into the impact of MCP-1 derived from BMAs on islet cells and its potential mechanisms, it is imperative to develop genetically engineered mice with conditional Mcp-1 knockout from BMAs.
Collapse
Affiliation(s)
- Shan Wan
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jinwei Xie
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Liang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
25
|
Orchanian SB, Lodoen MB. Monocytes as primary defenders against Toxoplasma gondii infection. Trends Parasitol 2023; 39:837-849. [PMID: 37633758 DOI: 10.1016/j.pt.2023.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/28/2023]
Abstract
Monocytes are recruited from the bone marrow to sites of infection where they release cytokines and chemokines, function in antimicrobial immunity, and differentiate into macrophages and dendritic cells to control infection. Although many studies have focused on monocyte-derived macrophages and dendritic cells, recent work has examined the unique roles of monocytes during infection to promote immune defense. We focus on the effector functions of monocytes during infection with the parasite Toxoplasma gondii, and discuss the signals that mobilize monocytes to sites of infection, their production of inflammatory cytokines and antimicrobial mediators, their ability to shape the adaptive immune response, and their immunoregulatory functions. Insights from other infections, including Plasmodium and Listeria are also included for comparison and context.
Collapse
Affiliation(s)
- Stephanie B Orchanian
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA; Institute for Immunology, University of California Irvine, Irvine, California, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA; Institute for Immunology, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
26
|
Tartara F, Montalbetti A, Crobeddu E, Armocida D, Tavazzi E, Cardia A, Cenzato M, Boeris D, Garbossa D, Cofano F. Compartmental Cerebrospinal Fluid Events Occurring after Subarachnoid Hemorrhage: An "Heparin Oriented" Systematic Review. Int J Mol Sci 2023; 24:7832. [PMID: 37175544 PMCID: PMC10178276 DOI: 10.3390/ijms24097832] [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: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a severe acute event with high morbidity and mortality due to the development of early brain injury (EBI), secondary delayed cerebral ischemia (DCI), and shunt-related hydrocephalus. Secondary events (SSE) such as neuroinflammation, vasospasm, excitotoxicity, blood-brain barrier disruption, oxidative cascade, and neuronal apoptosis are related to DCI. Despite improvement in management strategies and therapeutic protocols, surviving patients frequently present neurological deficits with neurocognitive impairment. The aim of this paper is to offer to clinicians a practical review of the actually documented pathophysiological events following subarachnoid hemorrhage. To reach our goal we performed a literature review analyzing reported studies regarding the mediators involved in the pathophysiological events following SAH occurring in the cerebrospinal fluid (CSF) (hemoglobin degradation products, platelets, complement, cytokines, chemokines, leucocytes, endothelin-1, NO-synthase, osteopontin, matricellular proteins, blood-brain barrier disruption, microglia polarization). The cascade of pathophysiological events secondary to SAH is very complex and involves several interconnected, but also distinct pathways. The identification of single therapeutical targets or specific pharmacological agents may be a limited strategy able to block only selective pathophysiological paths, but not the global evolution of SAH-related events. We report furthermore on the role of heparin in SAH management and discuss the rationale for use of intrathecal heparin as a pleiotropic therapeutical agent. The combination of the anticoagulant effect and the ability to interfere with SSE theoretically make heparin a very interesting molecule for SAH management.
Collapse
Affiliation(s)
- Fulvio Tartara
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Montalbetti
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Emanuela Crobeddu
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Daniele Armocida
- A.U.O. Policlinico Umberto I, Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00185 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Eleonora Tavazzi
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of Southern Switzerland, EOC, 6900 Lugano, Switzerland
| | - Marco Cenzato
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Davide Boeris
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Diego Garbossa
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| | - Fabio Cofano
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| |
Collapse
|
27
|
Lei W, Jia L, Wang Z, Liang Z, Aizhen Z, Liu Y, Tian Y, Zhao L, Chen Y, Shi G, Yang Z, Yang Y, Xu X. CC chemokines family in fibrosis and aging: From mechanisms to therapy. Ageing Res Rev 2023; 87:101900. [PMID: 36871782 DOI: 10.1016/j.arr.2023.101900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/21/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Fibrosis is a universal aging-related pathological process in the different organ, but is actually a self-repair excessive response. To date, it still remains a large unmet therapeutic need to restore injured tissue architecture without detrimental side effects, due to the limited clinical success in the treatment of fibrotic disease. Although specific organ fibrosis and the associated triggers have distinct pathophysiological and clinical manifestations, they often share involved cascades and common traits, including inflammatory stimuli, endothelial cell injury, and macrophage recruitment. These pathological processes can be widely controlled by a kind of cytokines, namely chemokines. Chemokines act as a potent chemoattractant to regulate cell trafficking, angiogenesis, and extracellular matrix (ECM). Based on the position and number of N-terminal cysteine residues, chemokines are divided into four groups: the CXC group, the CX3C group, the (X)C group, and the CC group. The CC chemokine classes (28 members) is the most numerous and diverse subfamily of the four chemokine groups. In this Review, we summarized the latest advances in the understanding of the importance of CC chemokine in the pathogenesis of fibrosis and aging and discussed potential clinical therapeutic strategies and perspectives aimed at resolving excessive scarring formation.
Collapse
Affiliation(s)
- Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Liyuan Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, 430064, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou 450052, China
| | - Zhao Aizhen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Yanqing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Ye Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yawu Chen
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Guangyong Shi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
28
|
Abstract
Tumour progression is modulated by the local microenvironment. This environment is populated by many immune cells, of which macrophages are among the most abundant. Clinical correlative data and a plethora of preclinical studies in mouse models of cancers have shown that tumour-associated macrophages (TAMs) play a cancer-promoting role. Within the primary tumour, TAMs promote tumour cell invasion and intravasation and tumour stem cell viability and induce angiogenesis. At the metastatic site, metastasis-associated macrophages promote extravasation, tumour cell survival and persistent growth, as well as maintain tumour cell dormancy in some contexts. In both the primary and metastatic sites, TAMs are suppressive to the activities of cytotoxic T and natural killer cells that have the potential to eradicate tumours. Such activities suggest that TAMs will be a major target for therapeutic intervention. In this Perspective article, we chronologically explore the evolution of our understanding of TAM biology put into the context of major enabling advances in macrophage biology.
Collapse
Affiliation(s)
| | - Jeffrey W Pollard
- MRC-Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
29
|
Kang J, Postigo-Fernandez J, Kim K, Zhu C, Yu J, Meroni M, Mayfield B, Bartolomé A, Dapito DH, Ferrante AW, Dongiovanni P, Valenti L, Creusot RJ, Pajvani UB. Notch-mediated hepatocyte MCP-1 secretion causes liver fibrosis. JCI Insight 2023; 8:e165369. [PMID: 36752206 PMCID: PMC9977430 DOI: 10.1172/jci.insight.165369] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/29/2022] [Indexed: 02/09/2023] Open
Abstract
Patients with nonalcoholic steatohepatitis (NASH) have increased expression of liver monocyte chemoattractant protein-1 (MCP-1), but its cellular source and contribution to various aspects of NASH pathophysiology remain debated. We demonstrated increased liver CCL2 (which encodes MCP-1) expression in patients with NASH, and commensurately, a 100-fold increase in hepatocyte Ccl2 expression in a mouse model of NASH, accompanied by increased liver monocyte-derived macrophage (MoMF) infiltrate and liver fibrosis. To test repercussions of increased hepatocyte-derived MCP-1, we generated hepatocyte-specific Ccl2-knockout mice, which showed reduced liver MoMF infiltrate as well as decreased liver fibrosis. Forced hepatocyte MCP-1 expression provoked the opposite phenotype in chow-fed wild-type mice. Consistent with increased hepatocyte Notch signaling in NASH, we observed a close correlation between markers of Notch activation and CCL2 expression in patients with NASH. We found that an evolutionarily conserved Notch/recombination signal binding protein for immunoglobulin kappa J region binding site in the Ccl2 promoter mediated transactivation of the Ccl2 promoter in NASH diet-fed mice. Increased liver MoMF infiltrate and liver fibrosis seen in opposite gain-of-function mice was ameliorated with concomitant hepatocyte Ccl2 knockout or CCR2 inhibitor treatment. Hepatocyte Notch activation prompts MCP-1-dependent increase in liver MoMF infiltration and fibrosis.
Collapse
Affiliation(s)
- Jinku Kang
- Department of Medicine, Naomi Berrie Diabetes Center, and
| | - Jorge Postigo-Fernandez
- Department of Medicine, Naomi Berrie Diabetes Center, and
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - KyeongJin Kim
- Department of Medicine, Naomi Berrie Diabetes Center, and
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science & Engineering, and Research Center for Controlling Intercellular Communication (RCIC), Inha University, Incheon, South Korea
| | - Changyu Zhu
- Department of Medicine, Naomi Berrie Diabetes Center, and
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Junjie Yu
- Department of Medicine, Naomi Berrie Diabetes Center, and
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Brent Mayfield
- Department of Medicine, Naomi Berrie Diabetes Center, and
| | - Alberto Bartolomé
- Department of Medicine, Naomi Berrie Diabetes Center, and
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | | | | | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine Lab, Biological Resource Center, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico Milan, Milan, Italy
| | - Remi J. Creusot
- Department of Medicine, Naomi Berrie Diabetes Center, and
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | | |
Collapse
|
30
|
Xu H, Yang F, Bao Z. Gut microbiota and myocardial fibrosis. Eur J Pharmacol 2023; 940:175355. [PMID: 36309048 DOI: 10.1016/j.ejphar.2022.175355] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 01/18/2023]
Abstract
Myocardial fibrosis (MF) is a pathophysiological condition that accompanies various myocardial diseases and comprises a damaged myocardial matrix repair process. Although fibrosis plays a vital role in repair, it ultimately alters cardiac systolic and diastolic functions. The gut microbiota is a complex and dynamic ecosystem with billions of microorganisms that produce bioactive compounds that influence host health and disease progression. Intestinal microbiota has been shown to correlate with cardiovascular disease, and dysbiosis of the intestinal microbiota is involved in the development of MF. In this review, we discuss the role of intestinal microbiota in the process of MF, including alterations in microbiota composition and the effects of metabolites. We also discuss how diet and medicines can affect cardiac fibrosis by influencing the gut microbiota, and potential future therapies targeting the gut-heart axis. A healthy gut microbiota can prevent disease, but dysbiosis can lead to various symptoms, including the induction of heart disease. In this review, we discuss the relevance of the gut-heart axis and the multiple pathways by which gut microbiota may affect cardiac fibrosis, including inflammatory factors, immune cells, and gut microbiota metabolites, such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs). Finally, we discuss the involvement of gut microbiota in the treatment of cardiac fibrosis, including drugs, fecal microbiota transplantation, and oral probiotics or prebiotics. With future studies on the relationship between the heart and gut microbiota, we hope to find better ways to improve MF through the gut-heart axis.
Collapse
Affiliation(s)
- Han Xu
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Zhang B, Hong L, Ke J, Zhong Y, Cao N, Li W, Xu D, Tian Y, Huang Y, Chen W, Li B. Polysaccharide of Atractylodes macrocephala Koidz alleviate lipopolysaccharide-induced liver injury in goslings via the p53 and FOXO pathways. Poult Sci 2023; 102:102480. [PMID: 36680857 PMCID: PMC9871332 DOI: 10.1016/j.psj.2023.102480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/13/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023] Open
Abstract
Lipopolysaccharide (LPS) can affect the immune system of geese by inducing liver injury. The polysaccharide of Atractylodes macrocephala Koidz (PAMK) have obvious immune-enhancing effects. Accordingly, this experiment investigated the effect of PAMK on LPS-induced liver injury in goslings. Two hundred 1-day-old goslings were randomly divided into the control group, LPS group, PAMK group, and PAMK+ LPS group, and the PAMK and PAMK+ LPS groups were fed the basal diet with 400 mg/kg PAMK, while the control and LPS groups were fed the basal diet. On D 21, 23, and 25 of the formal trial, the goslings in the LPS and PAMK+LPS groups were injected intraperitoneally with 2 mg/kg LPS, and goslings in the control and PAMK groups were injected intraperitoneally with the same amount of saline. Livers were collected on D 25. HE-stained sections showed that PAMK could effectively alleviate the LPS-induced indistinct hepatic cord structure, loss of cytoplasmic contents of hepatocytes, and dilatation of hepatic sinusoids. The biochemical parameters of liver tissues showed that PAMK could alleviate the LPS-induced upregulation of alanine aminotransferase and aspartate aminotransferase. To further investigate the mechanism of the mitigating effect of PAMK on LPS-induced injury, livers from the LPS and PAMK+LPS groups were selected for transcriptome sequencing. The sequencing results showed that there were 406 differentially expressed genes (DEGs) in the livers of LPS and PAMK+LPS goslings, of which 242 upregulated and 164 downregulated. The Kyoto Encyclopedia of Genes and Genome (KEGG) analysis showed that DEGs were significantly enriched in immune signal transduction, cell cycle, and cell metabolism. Besides, protein‒protein interaction analysis showed that 129 DEGs were associated with each other, including 7 DEGs enriched in the p53 and FOXO signaling pathway. In conclusion, PAMK may alleviate LPS-induced liver injury in gosling through the p53 and FOXO signaling pathway. These results provide a basis for further development of PAMK as an immunomodulator.
Collapse
Affiliation(s)
- Bingqi Zhang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Longsheng Hong
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Jingfei Ke
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Yueyun Zhong
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Nan Cao
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Wanyan Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Danning Xu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Yunbo Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Yunmao Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Wenbin Chen
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Bingxin Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China.
| |
Collapse
|
32
|
Mir MA, Bashir M, Ishfaq. Role of the CXCL8–CXCR1/2 Axis in Cancer and Inflammatory Diseases. CYTOKINE AND CHEMOKINE NETWORKS IN CANCER 2023:291-329. [DOI: 10.1007/978-981-99-4657-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Soliman AM, Barreda DR. Acute Inflammation in Tissue Healing. Int J Mol Sci 2022; 24:ijms24010641. [PMID: 36614083 PMCID: PMC9820461 DOI: 10.3390/ijms24010641] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
There are well-established links between acute inflammation and successful tissue repair across evolution. Innate immune reactions contribute significantly to pathogen clearance and activation of subsequent reparative events. A network of molecular and cellular regulators supports antimicrobial and tissue repair functions throughout the healing process. A delicate balance must be achieved between protection and the potential for collateral tissue damage associated with overt inflammation. In this review, we summarize the contributions of key cellular and molecular components to the acute inflammatory process and the effective and timely transition toward activation of tissue repair mechanisms. We further discuss how the disruption of inflammatory responses ultimately results in chronic non-healing injuries.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence: ; Tel.: +1-(780)492-0375
| |
Collapse
|
34
|
Disparate Regions of the Human Chemokine CXCL10 Exhibit Broad-Spectrum Antimicrobial Activity against Biodefense and Antibiotic-Resistant Bacterial Pathogens. ACS Infect Dis 2022; 9:122-139. [PMID: 36475632 PMCID: PMC9841529 DOI: 10.1021/acsinfecdis.2c00456] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CXCL10 is a pro-inflammatory chemokine produced by the host in response to microbial infection. In addition to canonical, receptor-dependent actions affecting immune-cell migration and activation, CXCL10 has also been found to directly kill a broad range of pathogenic bacteria. Prior investigations suggest that the bactericidal effects of CXCL10 occur through two distinct pathways that compromise the cell envelope. These observations raise the intriguing notion that CXCL10 features a separable pair of antimicrobial domains. Herein, we affirm this possibility through peptide-based mapping and structure/function analyses, which demonstrate that discrete peptides derived from the N- and C-terminal regions of CXCL10 mediate bacterial killing. The N-terminal derivative, peptide P1, exhibited marked antimicrobial activity against Bacillus anthracis vegetative bacilli and spores, as well as antibiotic-resistant clinical isolates of Klebsiella pneumoniae, Acinetobacter baumannii, Enterococcus faecium, and Staphylococcus aureus, among others. At bactericidal concentrations, peptide P1 had a minimal degree of chemotactic activity, but did not cause red blood cell hemolysis or cytotoxic effects against primary human cells. The C-terminal derivative, peptide P9, exhibited antimicrobial effects, but only against Gram-negative bacteria in low-salt medium─conditions under which the peptide can adopt an α-helical conformation. The introduction of a hydrocarbon staple induced and stabilized α-helicity; accordingly, stapled peptide P9 displayed significantly improved bactericidal effects against both Gram-positive and Gram-negative bacteria in media containing physiologic levels of salt. Together, our findings identify and characterize the antimicrobial regions of CXCL10 and functionalize these novel determinants as discrete peptides with potential therapeutic utility against difficult-to-treat pathogens.
Collapse
|
35
|
Connors JP, Stelzer JW, Garvin PM, Wellington IJ, Solovyova O. The Role of the Innate Immune System in Wear Debris-Induced Inflammatory Peri-Implant Osteolysis in Total Joint Arthroplasty. Bioengineering (Basel) 2022; 9:764. [PMID: 36550970 PMCID: PMC9774505 DOI: 10.3390/bioengineering9120764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Periprosthetic osteolysis remains a leading complication of total hip and knee arthroplasty, often resulting in aseptic loosening of the implant and necessitating revision surgery. Wear-induced particulate debris is the main cause initiating this destructive process. The purpose of this article is to review recent advances in understanding of how wear debris causes osteolysis, and emergent strategies for the avoidance and treatment of this disease. A strong activator of the peri-implant innate immune this debris-induced inflammatory cascade is dictated by macrophage secretion of TNF-α, IL-1, IL-6, and IL-8, and PGE2, leading to peri-implant bone resorption through activation of osteoclasts and inhibition of osteoblasts through several mechanisms, including the RANK/RANKL/OPG pathway. Therapeutic agents against proinflammatory mediators, such as those targeting tumor necrosis factor (TNF), osteoclasts, and sclerostin, have shown promise in reducing peri-implant osteolysis in vitro and in vivo; however, radiographic changes and clinical diagnosis often lag considerably behind the initiation of osteolysis, making timely treatment difficult. Considerable efforts are underway to develop such diagnostic tools, therapies, and identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- John Patrick Connors
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - John W Stelzer
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Patrick M Garvin
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Ian J Wellington
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Olga Solovyova
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| |
Collapse
|
36
|
Israr M, DeVoti JA, Papayannakos CJ, Bonagura VR. Role of chemokines in HPV-induced cancers. Semin Cancer Biol 2022; 87:170-183. [PMID: 36402301 DOI: 10.1016/j.semcancer.2022.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Human papillomaviruses (HPVs) cause cancers of the uterine cervix, oropharynx, anus, and vulvovaginal tract. Low-risk HPVs, such as HPV6 and 11, can also cause benign mucosal lesions including genital warts, and in patients with recurrent respiratory papillomatosis, lesions in the larynx, and on occasion, in the lungs. However, both high and less tumorigenic HPVs share a striking commonality in manipulating both innate and adaptive immune responses in HPV- infected keratinocytes, the natural host for HPV infection. In addition, immune/inflammatory cell infiltration into the tumor microenvironment influences cancer growth and prognosis, and this process is tightly regulated by different chemokines. Chemokines are small proteins and exert their biological effects by binding with G protein-coupled chemokine receptors (GPCRs) that are found on the surfaces of select target cells. Chemokines are not only involved in the establishment of a pro-tumorigenic microenvironment and organ-directed metastases but also involved in disease progression through enhancing tumor cell growth and proliferation. Therefore, having a solid grasp on chemokines and immune checkpoint modulators can help in the treatment of these cancers. In this review, we discuss the recent advances on the expression patterns and regulation of the main chemokines found in HPV-induced cancers, and their effects on both immune and non-immune cells in these lesions. Importantly, we also present the current knowledge of therapeutic interventions on the expression of specific chemokine and their receptors that have been shown to influence the development and progression of HPV-induced cancers.
Collapse
Affiliation(s)
- Mohd Israr
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - James A DeVoti
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Christopher J Papayannakos
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Vincent R Bonagura
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States; The Department of Pediatrics, The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.
| |
Collapse
|
37
|
Activation of the Chemokine Receptor CCR1 and Preferential Recruitment of Gαi Suppress RSV Replication: Implications for Developing Novel Respiratory Syncytial Virus Treatment Strategies. J Virol 2022; 96:e0130922. [PMID: 36317881 PMCID: PMC9682993 DOI: 10.1128/jvi.01309-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major pathogen that can cause acute respiratory infectious diseases of the upper and lower respiratory tract, especially in children, elderly individuals, and immunocompromised people. Generally, following viral infection, respiratory epithelial cells secrete cytokines and chemokines to recruit immune cells and initiate innate and/or adaptive immune responses. However, whether chemokines affect viral replication in nonimmune cells is rarely clear. In this study, we detected that chemokine CCL5 was highly expressed, while expression of its receptor, CCR1, was downregulated in respiratory epithelial cells after RSV infection. When we overexpressed CCR1 on respiratory epithelial cells in vivo or in vitro, viral load was significantly suppressed, which can be restored by the neutralizing antibody for CCR1. Interestingly, the antiviral effect of CCR1 was not related to type I interferon (IFN-I), apoptosis induction, or viral adhesion or entry inhibition. In contrast, it was related to the preferential recruitment and activation of the adaptor Gαi, which promoted inositol 1,4,5-triphosphate receptor type 3 (ITPR3) expression, leading to inhibited STAT3 phosphorylation; explicitly, phosphorylated STAT3 (p-STAT3) was verified to be among the important factors regulating the activity of HSP90, which has been previously reported to be a chaperone of RSV RNA polymerase. In summary, we are the first to reveal that CCR1 on the surface of nonimmune cells regulates RSV replication through a previously unknown mechanism that does not involve IFN-I induction. IMPORTANCE Our results revealed a novel mechanism by which RSV escapes innate immunity. That is, although it induces high CCL5 expression, RSV might attenuate the binding of CCL5 by downregulating the expression of CCR1 in respiratory epithelial cells to weaken the inhibitory effect of CCR1 on HSP90 activity and thereby facilitate RSV replication in nonimmune cells. This study provides a new target for the development of co-antiviral inhibitors against other components of the host and co-molecular chaperone/HSP90 and provides a scientific basis for the search for effective broad-spectrum antiviral drugs.
Collapse
|
38
|
Elevated G-CSF, IL8, and HGF in patients with definite Meniere's disease may indicate the role of NET formation in triggering autoimmunity and autoinflammation. Sci Rep 2022; 12:16309. [PMID: 36175465 PMCID: PMC9522806 DOI: 10.1038/s41598-022-20774-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022] Open
Abstract
The etiology and mechanism causing Meniere’s disease (MD) are not understood. The present study investigated the possible molecular mechanism of autoimmunity and autoinflammation associated with MD. Thirty-eight patients with definite MD and 39 normal volunteers were recruited, and 48 human cytokines/chemokines were quantified. In patients with MD pure tone audiograms, tympanograms and standard blood tests were performed. The mean hearing loss in the worse ear was 44.1 dB nHL. Compared to the referents, the concentrations of TNFα, IL1α, IL8, CTACK, MIP1α, MIP1β, G-CSF, and HGF in the sera of patients with MD were significantly elevated, while those of TRAIL and PDGFBB were significantly decreased. The area under the receiver operating characteristic curve (AUC) showed that G-CSF, MIP1α, and IL8 were above 0.8 and could be used to diagnose MD (p < 0.01), and the AUCs of CTACK and HGF were above 0.7 and acceptable to discriminate the MD group from the control group (p < 0.01). The revised AUCs (1 − AUC) of TRAIL and PDGFBB were above 0.7 and could also be used in the diagnosis of MD (p < 0.01). The linear regression showed significant correlations between MIP1α and GCSF, between IL2Rα and GCSF, between IL8 and HGF, between MIP1α and IL8, and between SCF and CTACK; there was a marginal linear association between IP10 and MIP1α. Linear regression also showed that there were significant age-related correlations of CTACK and MIG expression in the MD group (p < 0.01, ANOVA) but not in the control group. We hypothesize that G-CSF, IL8, and HGF, which are involved in the development of neutrophil extracellular traps (NETs) and through various mechanisms influence the functions of macrophages, lymphocytes, and dendritic cells, among others, are key players in the development of EH and MD and could be useful in elucidating the pathophysiological mechanisms leading to MD. Biomarkers identified in the present study may suggest that both autoimmune and autoinflammatory mechanisms are involved in MD. In the future, it will be valuable to develop a cost-effective method to detect G-CSF, IL8, HGF, CTACK, MIP1α, TRAIL, and PDGFBB in the serum of patient that have diagnostic relevance.
Collapse
|
39
|
Chang TT, Chen C, Chen JW. CCL7 as a novel inflammatory mediator in cardiovascular disease, diabetes mellitus, and kidney disease. Cardiovasc Diabetol 2022; 21:185. [PMID: 36109744 PMCID: PMC9479413 DOI: 10.1186/s12933-022-01626-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractChemokines are key components in the pathology of chronic diseases. Chemokine CC motif ligand 7 (CCL7) is believed to be associated with cardiovascular disease, diabetes mellitus, and kidney disease. CCL7 may play a role in inflammatory events by attracting macrophages and monocytes to further amplify inflammatory processes and contribute to disease progression. However, CCL7-specific pathological signaling pathways need to be further confirmed in these chronic diseases. Given the multiple redundancy system among chemokines and their receptors, further experimental and clinical studies are needed to clarify whether direct CCL7 inhibition mechanisms could be a promising therapeutic approach to attenuating the development of cardiovascular disease, diabetes mellitus, and kidney disease.
Collapse
|
40
|
Nengroo MA, Khan MA, Verma A, Datta D. Demystifying the CXCR4 conundrum in cancer biology: Beyond the surface signaling paradigm. Biochim Biophys Acta Rev Cancer 2022; 1877:188790. [PMID: 36058380 DOI: 10.1016/j.bbcan.2022.188790] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 10/14/2022]
Abstract
The oncogenic chemokine duo CXCR4-CXCL12/SDF-1 (C-X-C Receptor 4-C-X-C Ligand 12/ Stromal-derived factor 1) has been the topic of intense scientific disquisitions since Muller et al., in her ground-breaking research, described this axis as a critical determinant of organ-specific metastasis in breast cancer. Elevated CXCR4 levels correlate with distant metastases, poor prognosis, and unfavourable outcomes in most solid tumors. Therapeutic impediment of the axis in clinics with Food and Drug Administration (FDA) approved inhibitors like AMD3100 or Plerixafor yield dubious results, contrary to pre-clinical developments. Clinical trials entailing inhibition of CXCR7 (C-X-C Receptor 7), another convicted chemokine receptor that exhibits affinity for CXCL12, reveal outcomes analogous to that of CXCR4-CXCL12 axis blockade. Of note, the cellular CXCR4 knockout phenotype varies largely from that of inhibitor treatments. These shaky findings pique great curiosity to delve further into the realm of this infamous chemokine receptor to provide a probable explanation. A multitude of recent reports suggests the presence of an increased intracellular CXCR4 pool in various cancers, both cytoplasmic and nuclear. This intracellular CXCR4 protein reserve seems active as it correlates with vital tumor attributes, viz. prognosis, aggressiveness, metastasis, and disease-free survival. Diminishing this entire intracellular CXCR4 load apart from the surface signals looks encouraging from a therapeutic point of view. Transcending beyond the classically accepted concept of ligand-mediated surface signaling, this review sheds new light on plausible associations of intracellularly compartmentalised CXCR4 with various aspects of tumorigenesis. Besides, this review also puts forward a comprehensive account of CXCR4 regulation in different cancers.
Collapse
Affiliation(s)
- Mushtaq Ahmad Nengroo
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow-226031, India
| | - Muqtada Ali Khan
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow-226031, India
| | - Ayushi Verma
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow-226031, India
| | - Dipak Datta
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow-226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
41
|
Liao HR, Kao YY, Leu YL, Liu FC, Tseng CP. Larixol inhibits fMLP-induced superoxide anion production and chemotaxis by targeting the βγ subunit of Gi-protein of fMLP receptor in human neutrophils. Biochem Pharmacol 2022; 201:115091. [DOI: 10.1016/j.bcp.2022.115091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022]
|
42
|
Liu H, Li MJ, Zhang XN, Wang S, Li LX, Guo FF, Zeng T. N,N-dimethylformamide-induced acute liver damage is driven by the activation of NLRP3 inflammasome in liver macrophages of mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113609. [PMID: 35551047 DOI: 10.1016/j.ecoenv.2022.113609] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
N,N-dimethylformamide (DMF) is a non-negligible volatile hazardous material in indoor and outdoor environments. Although the hepatotoxicity of DMF has been well recognized, the underlying mechanisms remain unclear and prophylactic medicine is still lacking. Herein, we established a DMF-induced acute liver injury mouse model and investigated the underlying mechanisms focusing on oxidative stress and the nucleotide-binding domain and leucine-rich repeat receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome. DMF was found to induce oxidative stress, evidenced by the elevation of hepatic malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) adducts levels, and the decline of reduced glutathione (GSH) levels. However, neither N-acetyl cysteine (NAC) nor sulforaphane (SF) ameliorated the hepatoxicity induced by DMF in mice. Interestingly, DMF exposure led to focal necrosis of hepatocytes and NLRP3 inflammasome activation before the onset of obvious liver damage. In addition, DMF exposure induced infiltration and proinflammatory/M1 polarization of macrophages in mice livers. Furthermore, the inactivation of hepatic macrophages by GdCl3 significantly suppressed DMF-induced elevation of serum aminotransferase activities, neutrophile infiltration, and activation of NLRP3 inflammasome in mice liver. Collectively, these results suggest that DMF-induced acute hepatotoxicity may be attributed to the activation of NLRP3 inflammasome in liver macrophages, but not oxidative stress.
Collapse
Affiliation(s)
- Hong Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ming-Jun Li
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Tangshan Vocational&Technical College, Tangshan, Hebei 063000, China
| | - Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shuo Wang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong Province 252059, China
| | - Long-Xia Li
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fang-Fang Guo
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
43
|
Yan D, Li W, Liu Q, Yang K. Advances in Immune Microenvironment and Immunotherapy of Isocitrate Dehydrogenase Mutated Glioma. Front Immunol 2022; 13:914618. [PMID: 35769466 PMCID: PMC9234270 DOI: 10.3389/fimmu.2022.914618] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022] Open
Abstract
The tumor immune microenvironment and immunotherapy have become current important tumor research concerns. The unique immune microenvironment plays a crucial role in the malignant progression of isocitrate dehydrogenase (IDH) mutant gliomas. IDH mutations in glioma can inhibit tumor-associated immune system evasion of NK cell immune surveillance. Meanwhile, mutant IDH can inhibit classical and alternative complement pathways and directly inhibit T-cell responses by metabolizing isocitrate to D-2-Hydroxyglutaric acid (2-HG). IDH has shown clinically relevant efficacy as a potential target for immunotherapy. This article intends to summarize the research progress in the immunosuppressive microenvironment and immunotherapy of IDH-mutant glioma in recent years in an attempt to provide new ideas for the study of occurrence, progression, and treatment of IDH-mutant glioma.
Collapse
Affiliation(s)
- Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Weicheng Li
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Pharmacology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| | - Kun Yang
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| |
Collapse
|
44
|
An Update on the Chemokine System in the Development of NAFLD. Medicina (B Aires) 2022; 58:medicina58060761. [PMID: 35744024 PMCID: PMC9227560 DOI: 10.3390/medicina58060761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world. Sustained hepatic inflammation is a key driver of the transition from simple fatty liver to nonalcoholic steatohepatitis (NASH), the more aggressive form of NAFLD. Hepatic inflammation is orchestrated by chemokines, a family of chemoattractant cytokines that are produced by hepatocytes, Kupffer cells (liver resident macrophages), hepatic stellate cells, endothelial cells, and vascular smooth muscle cells. Over the last three decades, accumulating evidence from both clinical and experimental investigations demonstrated that chemokines and their receptors are increased in the livers of NAFLD patients and that CC chemokine ligand (CCL) 2 and CCL5 in particular play a pivotal role in inducing insulin resistance, steatosis, inflammation, and fibrosis in liver disease. Cenicriviroc (CVC), a dual antagonist of these chemokines’ receptors, CCR2 and CCR5, has been tested in clinical trials in patients with NASH-associated liver fibrosis. Additionally, recent studies revealed that other chemokines, such as CCL3, CCL25, CX3C chemokine ligand 1 (CX3CL1), CXC chemokine ligand 1 (CXCL1), and CXCL16, can also contribute to the pathogenesis of NAFLD. Here, we review recent updates on the roles of chemokines in the development of NAFLD and their blockade as a potential therapeutic approach.
Collapse
|
45
|
Rath M, Schwefel K, Malinverno M, Skowronek D, Leopoldi A, Pilz RA, Biedenweg D, Bekeschus S, Penninger JM, Dejana E, Felbor U. Contact-dependent signaling triggers tumor-like proliferation of CCM3 knockout endothelial cells in co-culture with wild-type cells. Cell Mol Life Sci 2022; 79:340. [PMID: 35661927 PMCID: PMC9166869 DOI: 10.1007/s00018-022-04355-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 12/18/2022]
Abstract
Cerebral cavernous malformations (CCM) are low-flow vascular lesions prone to cause severe hemorrhage-associated neurological complications. Pathogenic germline variants in CCM1, CCM2, or CCM3 can be identified in nearly 100% of CCM patients with a positive family history. In line with the concept that tumor-like mechanisms are involved in CCM formation and growth, we here demonstrate an abnormally increased proliferation rate of CCM3-deficient endothelial cells in co-culture with wild-type cells and in mosaic human iPSC-derived vascular organoids. The observation that NSC59984, an anticancer drug, blocked the abnormal proliferation of mutant endothelial cells further supports this intriguing concept. Fluorescence-activated cell sorting and RNA sequencing revealed that co-culture induces upregulation of proangiogenic chemokine genes in wild-type endothelial cells. Furthermore, genes known to be significantly downregulated in CCM3−/− endothelial cell mono-cultures were upregulated back to normal levels in co-culture with wild-type cells. These results support the hypothesis that wild-type ECs facilitate the formation of a niche that promotes abnormal proliferation of mutant ECs. Thus, targeting the cancer-like features of CCMs is a promising new direction for drug development.
Collapse
|
46
|
The Most Promising Biomarkers of Allogeneic Kidney Transplant Rejection. J Immunol Res 2022; 2022:6572338. [PMID: 35669103 PMCID: PMC9167141 DOI: 10.1155/2022/6572338] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/30/2022] [Indexed: 12/13/2022] Open
Abstract
Clinical transplantology is a constantly evolving field of medicine. Kidney transplantation has become standard clinical practice, and it has a significant impact on reducing mortality and improving the quality of life of patients. Allogenic transplantation induces an immune response, which may lead to the rejection of the transplanted organ. The gold standard for evaluating rejection of the transplanted kidney by the recipient's organism is a biopsy of this organ. However, due to the high invasiveness of this procedure, alternative diagnostic methods are being sought. Therefore, the biomarkers may play an essential predictive role in transplant rejection. A review of the most promising biomarkers for early diagnosis and prognosis prediction of allogenic kidney transplant rejection summarizes novel data on neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), C-X-C motif chemokine 10 (CXCL-10), cystatin C (CysC), osteopontin (OPN), and clusterin (CLU) and analyses the dynamics of changes of the biomarkers mentioned above in kidney diseases and the mechanism of rejection of the transplanted kidney.
Collapse
|
47
|
Goïta AA, Guenot D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers (Basel) 2022; 14:1810. [PMID: 35406582 PMCID: PMC8997717 DOI: 10.3390/cancers14071810] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common cancers, and diagnosis at late metastatic stages is the main cause of death related to this cancer. This progression to metastasis is complex and involves different molecules such as the chemokine CXCL12 and its two receptors CXCR4 and CXCR7. The high expression of receptors in CRC is often associated with a poor prognosis and aggressiveness of the tumor. The interaction of CXCL12 and its receptors activates signaling pathways that induce chemotaxis, proliferation, migration, and cell invasion. To this end, receptor inhibitors were developed, and their use in preclinical and clinical studies is ongoing. This review provides an overview of studies involving CXCR4 and CXCR7 in CRC with an update on their targeting in anti-cancer therapies.
Collapse
Affiliation(s)
| | - Dominique Guenot
- INSERM U1113/Unistra, IRFAC—Interface de Recherche Fondamentale et Appliquée en Cancérologie, 67200 Strasbourg, France;
| |
Collapse
|
48
|
Xiong X, Liao X, Qiu S, Xu H, Zhang S, Wang S, Ai J, Yang L. CXCL8 in Tumor Biology and Its Implications for Clinical Translation. Front Mol Biosci 2022; 9:723846. [PMID: 35372515 PMCID: PMC8965068 DOI: 10.3389/fmolb.2022.723846] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
The chemokine CXCL8 has been found to play an important role in tumor progression in recent years. CXCL8 activates multiple intracellular signaling pathways by binding to its receptors (CXCR1/2), and plays dual pro-tumorigenic roles in the tumor microenvironment (TME) including directly promoting tumor survival and affecting components of TME to indirectly facilitate tumor progression, which include facilitating tumor cell proliferation and epithelial-to-mesenchymal transition (EMT), pro-angiogenesis, and inhibit anti-tumor immunity. More recently, clinical trials indicate that CXCL8 can act as an independently predictive biomarker in patients receiving immune checkpoint inhibitions (ICIs) therapy. Preclinical studies also suggest that combined CXCL8 blockade and ICIs therapy can enhance the anti-tumor efficacy, and several clinical trials are being conducted to evaluate this therapy modality.
Collapse
Affiliation(s)
- Xingyu Xiong
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Xinyang Liao
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- Center of Biomedical Big Data, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Xu
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shiyu Zhang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Sheng Wang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Jianzhong Ai
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Jianzhong Ai, ; Lu Yang,
| | - Lu Yang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Jianzhong Ai, ; Lu Yang,
| |
Collapse
|
49
|
Doganyigit Z, Eroglu E, Akyuz E. Inflammatory mediators of cytokines and chemokines in sepsis: From bench to bedside. Hum Exp Toxicol 2022; 41:9603271221078871. [PMID: 35337213 DOI: 10.1177/09603271221078871] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background: Sepsis is a serious clinical condition characterized by damage to the immune system as a result of an uncontrolled response to infection. Septic patients show complications such as fever, cardiovascular shock, and/or systemic organ failure. Acute organ failure formed in sepsis mostly affects the respiratory and cardiovascular systems. In sepsis, responses including pro-inflammatory and anti-inflammatory processes in addition to the Toll-Like Receptor 4 (TLR4) signals leading to the release of inflammatory mediators have been suggested to be fundamental pathways in the pathophysiology of sepsis. Purpose: In this context, unregulated levels of sepsis-associated inflammatory mediators may increase the risk of mortality. In sepsis, infection-induced pathogens lead to a systemic inflammatory response. These systemic responses may contribute to septic shock and organ dysfunction. In the unfavorable clinical course of sepsis, an uncontrolled inflammatory response is observed. Accordingly, the mechanism of inflammatory mediators such as cytokines and chemokines in sepsis might increase. Neurotransmitters and gene regulators affect inflammatory mediators and control the inflammatory response. In this review, we aimed to show the new therapeutic targets in sepsis treatment with current studies. New clinical implications targeting inflammatory mediators in high mortality affected by the uncontrolled inflammatory response in sepsis can contribute to the understanding of the symptoms.
Collapse
Affiliation(s)
- Zuleyha Doganyigit
- Faculty of Medicine, Histology and Embryology, 162338Yozgat Bozok University, Yozgat, Turkey
| | - Ece Eroglu
- Faculty of Medicine, 162338Yozgat Bozok University Yozgat, Turkey
| | - Enes Akyuz
- Faculty of International Medicine, Department of Biophysics, 448249University of Health Sciences Istanbul, Turkey
| |
Collapse
|
50
|
Identification and mechanism of G protein-biased ligands for chemokine receptor CCR1. Nat Chem Biol 2022; 18:264-271. [PMID: 34949837 PMCID: PMC8885419 DOI: 10.1038/s41589-021-00918-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
Biased signaling of G protein-coupled receptors describes an ability of different ligands that preferentially activate an alternative downstream signaling pathway. In this work, we identified and characterized different N-terminal truncations of endogenous chemokine CCL15 as balanced or biased agonists targeting CCR1, and presented three cryogenic-electron microscopy structures of the CCR1-Gi complex in the ligand-free form or bound to different CCL15 truncations with a resolution of 2.6-2.9 Å, illustrating the structural basis of natural biased signaling that initiates an inflammation response. Complemented with pharmacological and computational studies, these structures revealed it was the conformational change of Tyr291 (Y2917.43) in CCR1 that triggered its polar network rearrangement in the orthosteric binding pocket and allosterically regulated the activation of β-arrestin signaling. Our structure of CCL15-bound CCR1 also exhibited a critical site for ligand binding distinct from many other chemokine-receptor complexes, providing new insights into the mode of chemokine recognition.
Collapse
|