1
|
Franke BU, Kummer K, Rose-John S, Lichtenthaler SF, Kress M. Shedding new light on BACE1-mediated modulation of IL-6 signaling: Implications for neural activity and synaptic plasticity in mice. Cytokine 2025; 190:156925. [PMID: 40184913 DOI: 10.1016/j.cyto.2025.156925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
The pleiotropic cytokine IL-6 regulates numerous processes in the body, including neuronal functions. IL-6 either binds to membrane-bound receptor (mIL-6R) and triggers signaling via heteromerization with the signal transducer gp130 (classical signaling), or binds to its soluble form (sIL-6R) to act on cells that do not express mIL-6R (trans-signaling). The ß-secretase BACE1 can cleave gp130 as well as IL-6R and we hypothesized that BACE1 may alter neuron activity and synaptic transmission via modulation of IL-6 signaling. We used multielectrode array (MEA) recordings to monitor electrical activity of neuronal networks in acute cerebellar slices as well as long-term potentiation (LTP) induced by high-frequency stimulation in the hippocampus and to assess how exposure to IL-6 affects these processes. A pharmacological approach was applied to elucidate the contribution of trans-signaling involving BACE1. Spontaneous neuronal activity in cerebellar slices significantly decreased upon perfusion with IL-6 but not LIF and recovered during wash out. BACE1 inhibitors verubecestat or AZD3839 abolished the inhibitory effects of IL-6. Furthermore, IL-6 and LIF reversibly inhibited LTP in hippocampal slices, and in contrast to cerebellar neurons, BACE1 inhibitors verubecestat or AZD3839 did not abolish the inhibitory effect of IL-6 on LTP. Interestingly, a dramatic rebound effect on excitatory postsynaptic potentials was observed with BACE1 inhibitor AZD3839 but not verubecestat during wash out. Our results support relevant and differential roles of IL-6, LIF and BACE1 in pathways modulating neuronal discharge activity in the cerebellum and the synaptic plasticity in the hippocampus, and a possible involvement of this interaction in deficits of memory and learning.
Collapse
Affiliation(s)
- Buket Ucar Franke
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, Kiel, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Immunol Allergy Clin North Am 2025; 45:223-249. [PMID: 40287170 DOI: 10.1016/j.iac.2025.01.010] [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] [Indexed: 04/29/2025]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
3
|
Allahyari Z, Lotfabadi NN, Zare F. The role of leukemia inhibitory factor in regulating angiogenesis-related gene expression in a mouse model of recurrent miscarriage. Placenta 2025; 165:91-101. [PMID: 40228388 DOI: 10.1016/j.placenta.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Recurrent miscarriage is an early pregnancy complication that affects approximately 1-3 % of pregnant couples. Leukemia Inhibitory Factor (LIF) plays an important role in various biological processes, including angiogenesis and pregnancy. This study aimed to evaluate the role of LIF in regulating angiogenesis-related genes in a mouse model of recurrent miscarriage. METHOD Female CBA/J mice mated with DBA/2J males were utilized as a miscarriage model. The study population was randomly assigned to three groups, normal group, mating female CBA/J mouse with male Balb/c without injection; miscarriage model control group with PBS injection; and the miscarriage group, in which LIF was injected. Following detection of a vaginal plug, mice were dissected on days 4, 7, and 14 of pregnancy. Uterine and placental tissues were collected to assess the expression of angiogenesis-related genes, including VEGF, PDGF, ANG1, FGF, and TGF-β, using real-time PCR. RESULT Data analysis revealed no significant differences in the expression of angiogenesis-related genes on days 4 and 7 of pregnancy compared with the control group. However, on day 14 of pregnancy, the expression of VEGF and TGF-β was significantly elevated in the miscarriage group receiving LIF compared to other groups (P = 0.03 and P = 0.04, respectively). The placental expression of the studied genes also exhibited a non-significant increase in the miscarriage group, with VEGF and TGF-β showing the most prominent increases, although these changes were not statistically significant. Correlation analysis between uterine and placental gene expression on day 14 revealed no significant association. CONCLUSION LIF regulates the uterine and placental expression of angiogenesis-related genes, particularly VEGF and TGF-β. These findings highlight the role of LIF in regulating angiogenesis-related gene expression and suggest that LIF could be a potential therapeutic candidate for improving pregnancy outcomes in cases of recurrent miscarriage.
Collapse
Affiliation(s)
- Zahra Allahyari
- Molecular Genetics, Faculty of Science, Science and Arts University, Yazd, Iran
| | | | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
4
|
Wang C, He T, Qin J, Jiao J, Ji F. The roles of immune factors in neurodevelopment. Front Cell Neurosci 2025; 19:1451889. [PMID: 40276707 PMCID: PMC12018394 DOI: 10.3389/fncel.2025.1451889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
The development of the nervous system is a highly complex process orchestrated by a multitude of factors, including various immune elements. These immune components play a dual role, not only regulating the immune response but also actively influencing brain development under both physiological and pathological conditions. The brain's immune barrier includes microglia in the brain parenchyma, which act as resident macrophages, astrocytes that support neuronal function and contribute to the inflammatory response, as well as circulating immune cells that reside at the brain's borders, including the choroid plexus, meninges, and perivascular spaces. Cytokines-soluble signaling molecules released by immune cells-play a crucial role in mediating communication between immune cells and the developing nervous system. Cytokines regulate processes such as neurogenesis, synaptic pruning, and inflammation, helping to shape the neural environment. Dysregulation of these immune cells, astrocytes, or cytokine signaling can lead to alterations in neurodevelopment, potentially contributing to neurodevelopmental abnormalities. This article reviews the central role of microglia, astrocytes, cytokines, and other immune factors in neurodevelopment, and explores how neuroinflammation can lead to the onset of neurodevelopmental disorders, shedding new light on their pathogenesis.
Collapse
Affiliation(s)
- Chong Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
5
|
D'Mello V, Mihailovic J, Ali S, Sanganahalli BG, Coman D, Hyder F, Fernando M, Mampilly A, Kannurpatti SS, Levison SW. Leukemia Inhibitory Factor as a late-stage treatment for delayed white matter loss in concussive head injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647435. [PMID: 40291675 PMCID: PMC12026900 DOI: 10.1101/2025.04.07.647435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Leukemia Inhibitory Factor (LIF) is an injury-induced cytokine that peaks 48 hours after a traumatic brain injury (TBI). Juvenile LIF haplodeficient mice exhibit desynchronized glial responses, increased neurodegeneration, decreased axonal conductivity and behavioral deficits after a concussive head injury. Given the necessity of LIF during the acute recovery phase after injury, we hypothesized that intranasal (IN) LIF treatment would prevent neurodegeneration when administered during the chronic recovery period from a mild TBI (mTBI). Methods Young adult male CD1 mice were subjected to a midline, closed-head frontal cortex injury using a flat metal impactor with a 3mm tip to induce a mTBI. In the 6-8 weeks post-mTBI, known to precede axonal atrophy in this mTBI model, two doses of 40 ng and 100 ng of LIF were administered twice daily, 5 days/week for two consecutive weeks. Sensorimotor functions were assessed at 4 and 8 weeks post mTBI, followed by ex-vivo brain magnetic resonance imaging at 9.4T and histopathology. Findings mTBI mice showed sensorimotor deficits at 4 weeks, which worsened by 8 weeks post-injury. IN-LIF treatment prevented the progressive sensorimotor loss seen in the vehicle-treated controls. Increased mean diffusivity (MD) and decreased fractional anisotropy (FA) were observed in the corpus callosum and prefrontal cortex of mTBI brains. In a dose-dependent manner, IN-LIF prevented the mTBI-induced MD increase and FA decrease. Histologically, there was significantly less astrogliosis, microgliosis and axonal injury in the IN-LIF treated mice vs. controls. Interpretation These results support the therapeutic potential of IN-LIF to reduce delayed neurodegeneration and improve neurological outcomes after mTBIs. Funding Supported by R21 NS125201, which was awarded to SWL, SK, and FH, and Rutgers Busch Biomedical Grant IRES 21-002946 to SWL and SK. Research in context Evidence before this study: Earlier studies had shown that LIF haplodeficient mice sustained worse outcomes after brain injury, which supported the hypothesis that LIF was an essential neuroprotective injury induced cytokine. Other studies had shown that acutely administered LIF was neuroprotective and glioprotective in mouse models of multiple sclerosis, neonatal hypoxia-ischemia and pediatric TBI. However, to date most pre-clinical studies for TBI have tested the efficacy of therapeutics delivered during the acute (primary) or sub-acute (secondary) recovery period. Few studies have focused on the mechanisms of delayed neurodegeneration (tertiary neurodegeneration) and therapeutics are entirely lacking. Therefore, we decided to test IN LIF during the chronic recovery period from TBI. With preliminary data, we submitted an NIH exploratory grant (R21) that was awarded to the senior investigators of this manuscript in October of 2021. That grant supported the majority of the studies contained in this submission.A pubmed search performed on Feb. 9th, 2025 using the search string "(traumatic brain injury) AND (axonal damage) AND (magnetic resonance imaging) AND intranasal AND neuroprotection" returned no references.Added value of this study: The standard of care for individuals who have sustained head injuries is to treat their symptoms. They are provided medications to reduce seizures, decrease anxiety, reduce depression and reduce pain and other symptoms. However, none of these medications will prevent tertiary neurodegeneration. Given the number of individuals who have sustained head injuries, new therapeutics, especially therapeutics that can be easily administered, are needed. With the Superbowl having just taken place, there is once again increasing concern that many of these athletes who have sustained head injuries during the course of their careers will go on to develop chronic traumatic encephalopathy, for which there is no treatment.The studies we described herein are innovative as no other group has evaluated any of the cytokines related to LIF for their neuroprotective properties for mTBI and certainly not during the tertiary injury period. Moreover, a Pubmed database search that covered the period from 1966 to 2025 reveals that only a handful of other studies have used intranasal delivery of any compound to treat TBI, and all of these studies administered their therapeutic within 6 hours after an injury. Developing a long-lasting, CNS-targeted therapeutic that can be delivered as a simple nose spray will have a lasting impact on clinical medicine. Our studies presage future clinical trials to assess the therapeutic efficacy of intranasal LIF for individuals who have sustained mild TBIs.
Collapse
|
6
|
Lee CK, Wang FT, Huang CH, Lin HJ, Chan WH. Effects and regulatory mechanisms of bisphenol a on the increases apoptosis and decreases differentiation potential in mouse embryonic stem cells. Toxicol Res (Camb) 2025; 14:tfaf043. [PMID: 40161257 PMCID: PMC11950671 DOI: 10.1093/toxres/tfaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/05/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
Bisphenol A has deleterious effects on reproductive, developmental, cell biological, and physiological functions. Here, we investigated the dosage effects of bisphenol A on the differentiation potential and apoptosis of mouse embryonic stem cells, and assessed some relevant regulatory mechanisms. Our results showed that bisphenol A at doses of 1-2 μmol/L triggers apoptotic processes without necrotic cell death in the ESC-B5 mouse embryonic stem cell line. No death effect was seen at treatment dosages of 0.5 μmol/L or less. Mechanistically, the application of 1-2 μmol/L bisphenol A directly increased the intracellular oxidative stress levels, significantly increased the cytoplasmic calcium and nitric oxide contents, decreased the mitochondrial membrane potential, activated caspases-9 and -3, and triggered programmed cell death. Interestingly, embryoid body formation assays showed that 0.5 μmol/L bisphenol A decreased the differentiation potential of ESC-B5 cells without inducing apoptotic processes. Together, our results indicate that treatment with 1-2 μmol/L bisphenol A induces apoptosis and triggers hazardous effects on the differentiation and developmental potential of mouse embryonic stem cells in vitro. These results provide important evidence that bisphenol A should be considered a potent cytotoxin that has dose-dependent impacts on differentiation and apoptosis in a mouse embryonic stem cell line.
Collapse
Affiliation(s)
- Cheng-Kai Lee
- Department of Obstetrics and Gynecology, Taoyuan General Hospital, Ministry of Health & Welfare, Zhongshan Road, Taoyuan District, Taoyuan City 33004, Taiwan
| | - Fu-Ting Wang
- Rehabilitation and Technical Aid Center, Taipei Veterans General Hospital, Section 2, Shipai Road, Beitou District, Taipei City 11217, Taiwan
| | - Chien-Hsun Huang
- Hungchi Gene IVF Center, Daxing West Road, Taoyuan District, Taoyuan City 330012, Taiwan
| | - Hsin-Ju Lin
- Department of Pathology, Lin Shin Hospital, Section 3, Huizhong Road, Nantun District, Taichung City 40867, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City 32023, Taiwan
| |
Collapse
|
7
|
Huo L, Liu X, Wang H. Leukemia Inhibitory Factor Attenuates Hypoxic-Ischemic White Matter Injury via NLRP3 Inflammasome Activity Suppressing Through the Nrf2/HO-1 Pathway. FRONT BIOSCI-LANDMRK 2025; 30:36630. [PMID: 40152399 DOI: 10.31083/fbl36630] [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: 12/27/2024] [Revised: 02/07/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Inhibiting neuroinflammatory damage is an effective strategy for treating preterm white matter injury (PWMI). Leukemia inhibitory factor (LIF) can ameliorate (HI) induced white matter injury; however, the neuroprotective effects and mechanisms of LIF remain unclear. This study aimed to determine whether NOD-like receptor thermal protein domain associated protein (NLRP3)-dependent pyroptosis is involved in PWMI pathogenesis. METHODS We established an in vitro oxygen-glucose deprivation (OGD) cell model and an in vivo HI induced brain white matter injury neonatal mouse model. RNA sequencing (RNA-seq) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses examined differentially expressed genes in oxygen-glucose deprivation/reoxygenation (OGD/R) challenged CTX TNA2 rat astrocytes. The changes and effects of proteins were confirmed in neonatal rats in vitro and in vivo. Cell viability assays, reactive oxygen species (ROS) assays, apoptosis assays, and immunoblot were used to confirm LIF-mediated its neuroprotective effect against HI-induced white matter injury in vitro. RESULTS RNA-seq and KEGG analyses indicated OGD/R enriched NLRP3 inflammasome-related genes (validated by in vitro and in vivo models), showing that NLRP3-dependent pyroptosis proteins (apoptosis-associated speck-like protein contain a CARD (ASC), NLRP3, active caspase 1, IL-1β, IL-18, and N-terminal fragment of gasdermin D (GSDMD-N)) were all increased by HI or OGD/R. LIF upregulated HO-1 expression by activating Nrf2 via the MAPK and Akt kinase pathways and significantly decreased OGD/R-induced ROS production. NLRP3-dependent pyroptosis proteins were suppressed in the LIF group compared with those in the OGD/R and HI groups. Zinc protophyrin, an HO-1 inhibitor, partially abolished LIF-mediated viability enhancement in rat astrocytes. CONCLUSION NLRP3-dependent pyroptosis plays a role in PWMI pathogenesis; moreover, LIF mitigates OGD/R-induced pyroptosis-dependent neurotoxicity by upregulating HO-1 expression in rat astrocytes.
Collapse
Affiliation(s)
- Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, 110004 Shenyang, Liaoning, China
| | - Xueyan Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, 110004 Shenyang, Liaoning, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, 110004 Shenyang, Liaoning, China
| |
Collapse
|
8
|
Rocha RF, Coimbra LD, Fontoura MA, Ribeiro G, Sotorilli GE, Gomes GF, Borin A, Felipe J, Slowikowski E, Greison WSS, Cunha TM, Marques PE, Vieira PMM, Marques RE. Usutu virus-induced meningoencephalitis in immunocompetent mice is characterized by the recruitment of mononuclear cells and a proinflammatory T helper 1 response. J Virol 2025; 99:e0172424. [PMID: 39907280 PMCID: PMC11915786 DOI: 10.1128/jvi.01724-24] [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: 10/04/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Usutu virus (USUV) is an arbovirus and has emerged as a potential cause of encephalitis in humans and other vertebrates. The increasing detection of USUV in mosquitoes and birds across Africa and Central Europe, along with the lack of specific treatments or vaccines for many encephalitic orthoflaviviruses, underscores the need for focused research. In this study, we developed a USUV infection model in immunocompetent C57BL/6 mice (8-12 weeks old) to characterize disease development and associated inflammatory mechanisms. Mice were intracranially infected with 104 PFU of USUV, leading to neurological symptoms such as hunched posture, paralysis, conjunctivitis, and eventual death by day 6 post-infection. Meningeal cell infiltration and microglia activation were most prevalent in mouse brains; however, neuronal loss was not observed at the peak of the disease, which coincided with increased viral load and leukocyte infiltration. The immune response in the brain was marked by the systematic recruitment and activation of macrophages, neutrophils, and T lymphocytes. A noticeable shift was seen in CD4+ T cells toward T helper 1 (Th1) polarization, which corroborates a massive increase in the expression of Th1-associated cytokines and chemokines at the peak of infection, indicative of an augmented proinflammatory state. Additionally, a rise in regulatory T cells was observed, peaking on day 6 post-infection. These findings highlight the dynamic nature of the host response to USUV infection, enhance our understanding of the disease pathogenesis, and address the scarcity of immunocompetent experimental models for the investigation of neglected emerging flaviviruses.IMPORTANCEMosquito-borne viruses, including USUV, are maintained in nature through complex cycles involving arthropod vectors and vertebrate hosts. A comprehensive understanding of USUV biology and host-pathogen interactions is crucial for developing effective treatments, which necessitates reliable experimental models (G. J. Sips, J. Wilschut, and J. M. Smit, Rev Med Virol 22:69-87, 2012, https://doi.org/10.1002/rmv.712; T. C. Pierson and M. S. Diamond, Nat Microbiol 5:796-812, 2020, https://doi.org/10.1038/s41564-020-0714-0). The establishment of a USUV infection model in immunocompetent adult mice brings new perspectives on the inflammatory component of viral encephalitis, which is difficult to study in mice lacking antiviral interferon responses. Moreover, USUV is an emerging viral disease lacking therapeutic and preventive measures. The interplay of USUV pathogenesis and the host's immune response indicates that lymphocytes and monocytes participate in USUV infection in this model and could be explored in search of treatments targeting immunopathogenic processes triggered by infection.
Collapse
Affiliation(s)
- Rebeca Froes Rocha
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Laís D Coimbra
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Marina A Fontoura
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Molecular and Morphofunctional Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Guilherme Ribeiro
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Giuliana Eboli Sotorilli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Giovanni F Gomes
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Alexandre Borin
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Jaqueline Felipe
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Emily Slowikowski
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Flanders, Belgium
| | - Wilias Silva Santos Greison
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
- Graduate Program in Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Pedro Elias Marques
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Flanders, Belgium
| | - Pedro M M Vieira
- Graduate Program in Genetics and Molecular Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| |
Collapse
|
9
|
Demirel KJ, Neves Guimaraes A, Demirel I. The Role of Caspase-1 and Caspase-4 in Modulating Gingival Epithelial Cell Responses to Aggregatibacter actinomycetemcomitans Infection. Pathogens 2025; 14:295. [PMID: 40137780 PMCID: PMC11945752 DOI: 10.3390/pathogens14030295] [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: 02/14/2025] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by bacterial infection and immune dysregulation. Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) is a key pathogen linked to disease progression. Caspase-1 and caspase-4 regulate inflammasome activation and cytokine release, yet their roles in gingival epithelial immunity remain unclear. The aim of this study was to elucidate the involvement of caspase-1 and caspase-4 in regulating the immune response to A. actinomycetemcomitans infection in gingival epithelial cells. Human gingival epithelial cells (Ca9-22) and caspase-1- and caspase-4-deficient cells were infected with A. actinomycetemcomitans for 24 h. Inflammatory mediator release was analyzed using Olink proteomics. Bacterial colonization and invasion were assessed using fluorescence-based assays and gentamicin protection assays. Caspase-1- and caspase-4-deficient cells showed significantly altered cytokine and chemokine profiles after infection with A. actinomycetemcomitans, showing reduced IL-17C and IL-18 release. We also found an increased release of TGF-α and LIF from caspase-4-deficient cells, along with elevated levels of the chemokines IL-8, CXCL9, and CXCL10. Additionally, both caspase-1- and caspase-4-deficient cells showed increased bacterial colonization and invasion, particularly in caspase-4-deficient cells. These findings suggest that caspase-1 and caspase-4 play distinct yet essential roles in gingival epithelial immunity, regulating cytokine release, barrier integrity, and defense against A. actinomycetemcomitans colonization.
Collapse
Affiliation(s)
- Kartheyaene Jayaprakash Demirel
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden;
| | - Alessandra Neves Guimaraes
- Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden;
- Department of Periodontology and Implantology, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden;
| |
Collapse
|
10
|
Ramundo MS, da Fonseca GC, Ten-Caten F, Gerber AL, Guimarães AP, Manuli ER, Côrtes MF, Pereira GM, Brustolini O, Cabral MG, Dos Santos Lázari C, Brasil P, da Silveira Bressan C, Nakaya HI, Paranhos-Baccalà G, Vasconcelos ATR, Sabino EC. Transcriptomic insights into early mechanisms underlying post-chikungunya chronic inflammatory joint disease. Sci Rep 2025; 15:6745. [PMID: 40000671 PMCID: PMC11861634 DOI: 10.1038/s41598-025-86761-x] [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: 09/24/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Chikungunya virus (CHIKV) infection often results in a chronic joint condition known as Post-Chikungunya Chronic Inflammatory Joint Disease (pCHIKV-CIJD). This condition disrupts individuals' daily lives and contributes to increased healthcare expenditure. This study investigated the molecular mechanisms underlying pCHIKV-CIJD development by analyzing RNA transcripts, including small RNAs, of whole blood from CHIKV-infected patients. By comparing patients who evolved to pCHIKV-CIJD with those who did not, we identified molecular signatures associated with chronification in acute and post-acute disease phases. These molecules were primarily associated with an altered immune response regulation. Notably, LIFR, an immune receptor that enhanced IL-6 transcription, was down-regulated in the acute phase of pCHIKV-CIJD patients, while its inhibitor, hsa-miR-98-5p, was up-regulated in these individuals. Other downregulated genes include members of immune mechanisms whose impairment can lead to a reduction in the first line of antiviral response, thereby promoting virus persistence for a longer period in these patients. Additionally, pCHIKV-CIJD patients exhibited reduced transcript levels of MMP8, LFT, and DDIT4, genes already implicated in the pathological process of other types of inflammatory arthritis and seemingly relevant for pCHIKV-CIJD development. Overall, our findings provide insights into the early molecular mechanisms involved in the chronification and highlight potential targets for further investigation.
Collapse
Affiliation(s)
- Mariana Severo Ramundo
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
- Laboratório de Imunologia, LIM-19, Instituto do Coração (INCOR), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil, 05403-900.
| | | | - Felipe Ten-Caten
- Departamento de Moléstias Infecciosas e Parasitárias e Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
- Pathology Advanced Translational Research Unit (PATRU), Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandra L Gerber
- LABINFO, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Ana Paula Guimarães
- LABINFO, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Erika Regina Manuli
- Departamento de Moléstias Infecciosas e Parasitárias e Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
- Laboratorio de Investigaçao Medica LIM-46, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
- Universidade Municipal de São Caetano do Sul, São Caetano do Sul, Brazil
| | - Marina Farrel Côrtes
- Departamento de Moléstias Infecciosas e Parasitárias e Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Geovana Maria Pereira
- Departamento de Moléstias Infecciosas e Parasitárias e Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Otavio Brustolini
- LABINFO, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Milena Gomes Cabral
- Departamento de Moléstias Infecciosas e Parasitárias e Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Carolina Dos Santos Lázari
- Fleury Medicina e Saúde, São Paulo, Brazil
- Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Patrícia Brasil
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | | - Helder I Nakaya
- Scientific Platform Pasteur, Universidade de São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Instituto Todos Pela Saúde, São Paulo, Brazil
| | | | | | - Ester Cerdeira Sabino
- Laboratorio de Investigaçao Medica LIM-46, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
- Universidade Municipal de São Caetano do Sul, São Caetano do Sul, Brazil
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Romo B, Fuentes Z, Randolph L, Mahajan M, Aller EJ, Ebrahimi B, Santhamma B, Pratap UP, Subbarayalu P, Nagandla H, Thomas C, Nair HB, Vadlamudi RK, Viswanadhapalli S. Targeting the Leukemia Inhibitory Factor/Leukemia Inhibitory Factor Receptor Axis Reduces the Growth of Inflammatory Breast Cancer by Promoting Ferroptosis. Cancers (Basel) 2025; 17:790. [PMID: 40075639 PMCID: PMC11898489 DOI: 10.3390/cancers17050790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Inflammatory breast cancer (IBC) is a rare subtype of breast cancer accounting for 7% of breast cancer-related fatalities. There is an urgent need to develop new targeted treatments for IBC. The progression of IBC has been associated with alterations in growth factor and cytokine signaling; however, the function of the LIF (leukemia inhibitory factor)/LIFR (leukemia inhibitory factor receptor) cytokine pathway in the progression of IBC remains unknown. This study evaluated the role of LIFR signaling and tested the efficacy of the LIFR inhibitor EC359 in treating IBC. Methods: The utility of using LIFR inhibition as a treatment strategy in IBC was tested using cell survival, apoptosis, colony formation, invasion, and pre-clinical KPL4 xenografts. Western blotting, siRNA, RT-qPCR, and lipid peroxidation assays were used to establish the mechanism of EC359 therapy. Results: The reduction in LIFR levels using siRNA markedly decreased growth in colony formation assays and reduced the invasion of IBC cells. Pharmacological inhibition of LIFR with EC359 effectively reduced cell survival and the clonogenic capacity of IBC cells. RT-qPCR assays revealed that EC359 markedly decreased the expression of the LIFR target genes. Western blot analyses confirmed that EC359 treatment suppressed downstream LIF/LIFR signaling pathways and promoted apoptosis. Treatment of cells with the ferroptosis inhibitor Fer-1 negated the capacity of EC359 to induce apoptosis. Mechanistic investigations demonstrated that EC359 predominantly triggered ferroptosis by inhibiting the glutathione antioxidant defense system through the downregulation of Glutathione peroxidase 4 (GPX4) levels. EC359 (5 mg/kg/day) was effective in reducing the growth of the IBC KPL4 xenograft tumors. Conclusion: These findings demonstrates that LIFR inhibition promote ferroptosis-mediated cell death in IBC and that EC359 represent novel therapeutic for IBC treatment.
Collapse
Affiliation(s)
- Bianca Romo
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Zenaida Fuentes
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Lois Randolph
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Megharani Mahajan
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Emily J. Aller
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Behnam Ebrahimi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | | | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Panneerdoss Subbarayalu
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
- Greehey Children Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Harika Nagandla
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.N.); (C.T.)
| | - Christoforos Thomas
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.N.); (C.T.)
| | - Hareesh B. Nair
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Evestra, Inc., San Antonio, TX 78245, USA;
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| |
Collapse
|
12
|
Ferreira Dos Santos TC, Silva EN, Frezarim GB, Salatta BM, Baldi F, Simielli Fonseca LF, De Albuquerque LG, Magalhães Muniz MM, Dos Santos Silva DB. Cis-eQTL analysis reveals genes involved in biological processes of the immune system in Nelore cattle. Gene 2025; 937:149138. [PMID: 39638014 DOI: 10.1016/j.gene.2024.149138] [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: 08/27/2024] [Revised: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
The combination of transcriptional profiling and genotype data analyses enables the identification of genetic variants that may affect gene expression (eQTL - expression quantitative trait loci). This study aimed to identify cis-eQTL in Nellore cattle muscle tissue and determine their biological processes related to the immune system and involved eGenes. Genotypic data (SNP-Chip) and gene expression data (RNA-Seq) from a commercial population of 80 Nellore animals were evaluated. For the cis-eQTL identification, association tests were conducted for all variants near the gene (cis variants), followed by permutation tests to correct for multiple comparisons. Our analyses revealed 828 top cis-eQTL related to 1,062 genes of which most of these variants were in intronic and intergenic regions. The eQTLs rs109525554, rs109589165, rs110192253, rs133127698, rs137742430, rs41803313, rs43366333, and rs43711242 were associated with susceptibility and resistance to infections in cattle. Additionally, interferon family eGenes, such as IFNT3, IFN-TAU, IFNK, FYN, and IFNW1, and endothelial leukocyte migration, such as PRKCG and CXCL10 were found. These eGene families were linked to biological processes of innate and adaptive immune responses and associated with somatic cell scores in cattle, respectively. Our results may have implications for selecting desirable resistance traits in animals bred for production and highlight the importance of studying genetic variants involved in quantitative traits to improve our understanding of genetic mechanisms underlying gene expression regulation of adaptive traits in cattle.
Collapse
Affiliation(s)
- Thaís Cristina Ferreira Dos Santos
- University José do Rosário Vellano (UNIFENAS), Alfenas, Minas Gerais, Brazil; Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil.
| | - Evandro Neves Silva
- University José do Rosário Vellano (UNIFENAS), Alfenas, Minas Gerais, Brazil; Universidade Federal de Alfenas (UNIFAL), Alfenas, Minas Gerais, Brazil
| | | | - Bruna Maria Salatta
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, São Paulo, Brazil
| | - Fernando Baldi
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, São Paulo, Brazil
| | | | - Lucia Galvão De Albuquerque
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, São Paulo, Brazil; Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brasília, Federal District, Brazil
| | - Maria Malane Magalhães Muniz
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, São Paulo, Brazil; University of Guelph, UOGELPH, Canada
| | - Danielly Beraldo Dos Santos Silva
- University José do Rosário Vellano (UNIFENAS), Alfenas, Minas Gerais, Brazil; Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, São Paulo, Brazil.
| |
Collapse
|
13
|
Wang Z, Gu Y, Qu Y, Huang X, Sun T, Wu W, Hu Q, Chen X, Li Y, Zhao H, Hu Y, Wu B, Xu J. Prevention of Intrauterine Adhesion with Platelet-Rich Plasma Double-Network Hydrogel. Adv Biol (Weinh) 2025; 9:e2400336. [PMID: 39673358 DOI: 10.1002/adbi.202400336] [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: 06/16/2024] [Revised: 10/30/2024] [Indexed: 12/16/2024]
Abstract
Intrauterine adhesion (IUA) can negatively impact on pregnancy outcomes, leading to reduced pregnancy rates, secondary infertility, and an increased risk of pregnancy complications. Studies have shown that the application of platelet-rich plasma (PRP) in IUA patients is effective. However, the clinical readhesive rate of IUA after treatment is still high, especially in severe cases. Platelet-rich plasma double-network hydrogel (DN gel) is an engineered PRP double network hydrogel, which is a sodium alginate (SA) based PRP hydrogel with egg carton ion cross-linking and fibrin double network. The results of this study show that intrauterine injection of DN gel has a better effect on promoting endometrial regeneration and enhancing endometrial receptivity than PRP gel. The mechanism is analyzed through single-cell sequencing, which may be achieved by increasing the expression of neutrophils (Neut), natural killer cells (NK), and type I classical dendritic cells (cDC1) in the endometrium and inhibiting the infiltration of M2 macrophages. Overall, based on the good healing efficiency and good biocompatibility of DN gel, it is expected to become a method of treating IUA with better efficacy and faster clinical translation.
Collapse
Affiliation(s)
- Zhuomin Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ying Gu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiyuan Qu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xujia Huang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Tao Sun
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Department of Assisted Reproduction, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Qianyu Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xiao Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yu Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Huafei Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Bingbing Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jian Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Department of Assisted Reproduction, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
14
|
Gao B, Liu H, Zhu M, Zhang S, Wang M, Ruan Y, Zheng Y. Molecular dynamics simulations reveal key roles of the LIF receptor in the assembly of human LIF signaling complex. Comput Struct Biotechnol J 2025; 27:585-594. [PMID: 39989618 PMCID: PMC11847480 DOI: 10.1016/j.csbj.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
Leukemia inhibitory factor (LIF) is a critical cytokine involved in various biological processes, including stem cell self-renewal, inflammation, and cancer progression. Structural studies have revealed how LIF forms a functional signaling complex. However, the dynamic binding pattern of the complex remains inadequately clarified. In this study, we employed molecular dynamics (MD) simulations to investigate the recognition and binding mechanisms of LIF, revealing a preferential affinity for LIF Receptor (LIFR) over gp130, attributable to a larger buried surface area at the LIF-LIFR interface. Key residues F178 and K181 in FXXK motif, along with K124 in LIF helix B, mediate hydrophobic interactions, hydrogen bonding and allosteric regulation, collectively stabilizing the LIF-LIFR interaction. We propose that the unique N-terminal extension of LIF enables signaling without requiring the additional receptor subunit beyond gp130 and LIFR, as verified by cell proliferation assays, distinguishing it from other cytokines in the LIF family. Additionally, analysis of domain fluctuations revealed that the LIF-LIFR interface undergoes less angular displacement compared to the LIF-gp130 interface, indicating a more stable interaction with LIFR. Together, these findings provide valuable insights into the molecular basis of LIF recognition and binding, offering a dynamic foundation for cytokine engineering.
Collapse
Affiliation(s)
- Bo Gao
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- BGI Research, Hangzhou 310030, China
| | | | | | | | | | - Yijun Ruan
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yue Zheng
- BGI Research, Hangzhou 310030, China
- BGI Research, Changzhou 213299, China
| |
Collapse
|
15
|
Lv H, Lu K, Wang X, Zhang Y, Zhuang M, Li J, Su K, Gao W. Recent trends in research on the role of cholesterol in leukemia: a bibliometric and visualization study. Front Immunol 2025; 16:1511827. [PMID: 39917295 PMCID: PMC11799240 DOI: 10.3389/fimmu.2025.1511827] [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: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025] Open
Abstract
Background Cholesterol metabolism significantly impacts leukemia pathophysiology, affecting tumor cell survival, proliferation, and treatment resistance. This study employs bibliometric analysis and visualization techniques to investigate research trends regarding cholesterol in leukemia and identify key hotspots. Methods A systematic search of the Web of Science Core Collection was performed for literature published from 1980 to 2024 using the keywords "cholesterol" and "leukemia," yielding 1,220 articles. Bibliometric tools like VOSviewer and CiteSpace were utilized for visualizing citation networks and thematic clusters. Results The analysis comprised 1,220 publications produced by 6,771 researchers across 1,756 institutions in 68 countries, published in 576 journals with 5,903 unique keywords. Publication output demonstrated a significant rise from 1980 to 2024, peaking in 2022. The United States led in total publications (381) and citations (40,462), followed by China (137 articles) and Japan (102). Notably, U.S. publications had lower average citations than those from Germany and Brazil. Key institutions included the University of São Paulo, Medical College of Wisconsin, and National Cancer Institute, with prominent authors such as Maranhao Raul C. and Girotti Albert W. The journal Cancer Research was the most prolific, while Blood had the highest citation frequency. Major research areas encompassed molecular biology, immunology, and medicine, focusing on the cholesterol-leukemia link. Keyword co-occurrence and co-citation analyses reveal increasing interest in topics like STAT3, multidrug resistance, and treatment interactions. These insights suggest crucial areas for further research. Discussion Our findings emphasize cholesterol's significance in leukemia, indicating its potential as a therapeutic target. Further exploration at the intersection of cholesterol metabolism and leukemia requires multidisciplinary collaboration. Conclusion This bibliometric study delineates the evolving research landscape on cholesterol's role in leukemia, pinpointing emerging trends and future research directions to inform effective therapeutic strategies.
Collapse
Affiliation(s)
- Huijuan Lv
- Department of Oncology, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| | - Ke Lu
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ximing Wang
- Department of Surgery, The Second People’s Hospital of Jinan, Jinan, Shandong, China
| | - Yanfang Zhang
- Department of Laboratory, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| | - Mengqi Zhuang
- Department of Oncology, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| | - Jing Li
- Department of Oncology, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| | - Keli Su
- Department of Oncology, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| | - Wen Gao
- Department of Cardiology, Fourth People’s Hospital of Jinan City, Jinan, Shandong, China
| |
Collapse
|
16
|
Shang Y, Zhang Q, Ding Y, Wang Y, Gu S, Zang X, Xu Z, Huang S, Li Z, Wu Z, Gu T, Hong L, Cai G. The Expression Pattern and Functional Analysis of Extracellular Vesicle Long Non-Coding RNAs from Uterine Fluid During Implantation in Pig. Animals (Basel) 2025; 15:245. [PMID: 39858245 PMCID: PMC11758334 DOI: 10.3390/ani15020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) act as competing endogenous RNAs and play significant roles in porcine embryo development. Extracellular vesicles (EVs) in the uterine fluid (UF) can target and deliver maternal endometrial signalling molecules to embryonic trophoblast cells, exerting crucial regulatory effects during embryo implantation. However, the specific roles of lncRNAs carried by UF-EVs during the embryo implantation period have not been thoroughly reported in the literature. In the present study, high-throughput sequencing and biological tools were applied to analyse lncRNAs in UF-EVs on days 9, 12, and 15 of pregnancy to identify key regulatory lncRNAs in UF-EVs during the porcine embryonic implantation period and to explore their expression patterns and functional roles. A total of 30,203 lncRNAs were identified and 7879 differentially expressed lncRNAs were screened, and qRT-PCR was used to verify the sequencing data. Days 9-12 of pregnancy represent a critical stage of embryo implantation characterised by substantial morphological changes in porcine embryos. During this period, we identified a total of 4348 differentially expressed lncRNAs. Through screening and validation, we discovered that LNC_026212 was highly expressed on day 12 of pregnancy and can promote the proliferation and migration of porcine trophoblast cells (PTr cells). These novel findings contribute to our understanding of the impact of lncRNAs on porcine reproductive processes, offering new research directions to improve the success rate of embryo implantation in pigs.
Collapse
Affiliation(s)
- Yijun Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Qiuping Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yue Ding
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yongzhong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Shengchen Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Xupeng Zang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zheng Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Sixiu Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zicong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Ting Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| |
Collapse
|
17
|
Ma W, Yan H, Ma H, Xu Z, Dai W, Wu Y, Zhang H, Li Y. Roles of leukemia inhibitory factor receptor in cancer. Int J Cancer 2025; 156:262-273. [PMID: 39279155 DOI: 10.1002/ijc.35157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/19/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Leukemia inhibitory factor receptor (LIFR), in complex with glycoprotein 130 (gp130) as the receptor for leukemia inhibitory factor (LIF), can bind to a variety of cytokines and subsequently activate a variety of signaling pathways, including Janus kinase/signal transducer and activator of transcription 3. LIF, the most multifunctional cytokines of the interleukin-6 family acts as both a growth factor and a growth inhibitor in different types of tumors. LIF/LIFR signaling regulates a broad array of tumor-related processes including proliferation, apoptosis, migration, invasion. However, due to the activation of different signaling pathways, opposite regulatory effects are observed in certain tumor cells. Therefore, the role of LIFR in human cancers varies across different tumor and tissue, despite their recognized value in tumor treatment and prognosis observation is affirmed. Given its aberrant expression in numerous tumor cells and crucial regulatory function in tumorigenesis and progression, LIFR is considered as a promising targeted therapeutic agent. This review provides an overview of LIFR's initiating signaling pathway function as a cytokine receptor and summarize the current literature on the role of LIFR in cancer and its possible use in therapy.
Collapse
Affiliation(s)
- Wei Ma
- School of Stomatology, China Medical University, Shenyang, China
| | - Haixu Yan
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Haoyuan Ma
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Zengyan Xu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Wei Dai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yudan Wu
- School of Nursing, China Medical University, Shenyang, China
| | - Hongyan Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yanshu Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
18
|
Ye J, Qin SS, Hughson AL, Hannon G, Vrooman TG, Lesch ML, Eckl SL, Benoodt L, Mills BN, Lord EM, Belt BA, Linehan DC, Luheshi N, Eyles J, Gerber SA. Blockade of LIF and PD-L1 Enhances Chemotherapy in Preclinical PDAC Models. Cancers (Basel) 2025; 17:204. [PMID: 39857986 PMCID: PMC11763610 DOI: 10.3390/cancers17020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC), expecting to be the second leading cause of cancer deaths by 2030, resists immune checkpoint therapies due to its immunosuppressive tumor microenvironment (TME). Leukemia inhibitory factor (LIF) is a key target in PDAC, promoting stemness, epithelial-mesenchymal transition (EMT), and therapy resistance. Phase 1 clinical trials showed anti-LIF therapy is safe but with limited efficacy, suggesting better outcomes when combined with chemotherapy, radiotherapy, or immunotherapy. Methods: We assessed the combination of chemotherapy (gemcitabine/nab-paclitaxel) and dual blockade of LIF and PD-L1 on tumor growth and survival in orthotopic and spontaneous PDAC models. Flow cytometry and scRNA-seq were utilized to monitor the antitumor immune response. The role of key immune cells was further confirmed by depleting these immune cells, including CD4, CD8, or inflammatory monocytes. Results: Sequential treatment with chemotherapy (gemcitabine/nab-paclitaxel) and dual blockade of LIF and PD-L1 significantly improved antitumor efficacy compared to monotherapy or dual combinations of these therapies. This chemo/anti-LIF/anti-PD-L1 approach reduced EMT in tumor cells and enhanced the antitumor immune response, primarily through CD8 T cells, as depleting CD8 cells largely abrogated the effect of treatment. This combination therapy also shifted macrophages and dendritic cells towards an antitumor phenotype. Conclusions: The combination of chemotherapy, anti-LIF, and anti-PD-L1 not only targeted tumor cells but also augmented the anti-tumor immune response. These findings strongly support advancing chemo/anti-LIF/anti-PD-L1 combination therapy to clinical trials in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Shuyang S. Qin
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Angela L. Hughson
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Tara G. Vrooman
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sarah L. Eckl
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lauren Benoodt
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Bradley N. Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Edith M. Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brian A. Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - David C. Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Nadia Luheshi
- Oncology R&D, AstraZeneca, Aaron Klug Building, Granta Park, Cambridge CB2 0AA, UK; (N.L.); (J.E.)
| | - Jim Eyles
- Oncology R&D, AstraZeneca, Aaron Klug Building, Granta Park, Cambridge CB2 0AA, UK; (N.L.); (J.E.)
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14626, USA
| |
Collapse
|
19
|
Maggs LR, McVey M. REV7: a small but mighty regulator of genome maintenance and cancer development. Front Oncol 2025; 14:1516165. [PMID: 39839778 PMCID: PMC11747621 DOI: 10.3389/fonc.2024.1516165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
REV7, also known as MAD2B, MAD2L2, and FANCV, is a HORMA-domain family protein crucial to multiple genome stability pathways. REV7's canonical role is as a member of polymerase ζ, a specialized translesion synthesis polymerase essential for DNA damage tolerance. REV7 also ensures accurate cell cycle progression and prevents premature mitotic progression by sequestering an anaphase-promoting complex/cyclosome activator. Additionally, REV7 supports genome integrity by directing double-strand break repair pathway choice as part of the recently characterized mammalian shieldin complex. Given that genome instability is a hallmark of cancer, it is unsurprising that REV7, with its numerous genome maintenance roles, is implicated in multiple malignancies, including ovarian cancer, glioma, breast cancer, malignant melanoma, and small-cell lung cancer. Moreover, high REV7 expression is associated with poor prognoses and treatment resistance in these and other cancers. Promisingly, early studies indicate that REV7 suppression enhances sensitivity to chemotherapeutics, including cisplatin. This review aims to provide a comprehensive overview of REV7's myriad roles in genome maintenance and other functions as well as offer an updated summary of its connections to cancer and treatment resistance.
Collapse
Affiliation(s)
- Lara R. Maggs
- Department of Biology, Tufts University, Medford, MA, United States
| | - Mitch McVey
- Department of Biology, Tufts University, Medford, MA, United States
| |
Collapse
|
20
|
Yu J, Liang P. A Mendelian randomization study on associations between plasma lipidome, circulating inflammatory proteins, and erectile dysfunction. Transl Androl Urol 2024; 13:2724-2734. [PMID: 39816232 PMCID: PMC11732301 DOI: 10.21037/tau-24-378] [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: 07/30/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025] Open
Abstract
Background Some studies suggest a potential association between plasma lipidome and erectile dysfunction (ED), but the underlying mechanism and whether circulating inflammatory proteins act as mediators remain unclear. The purpose of this study was to investigate the potential causal relationships between plasma lipidome, inflammatory proteins, and ED. Methods Plasma lipidome, circulating inflammatory proteins, and ED cases were identified based on the summary data from several large-scale genome-wide association studies (GWAS). The causal relationships of plasma lipidome and circulating inflammatory proteins with ED were explored by a bidirectional two-sample, two-sample Mendelian randomization (MR) method. The inverse variance weighted (IVW) method was used as the primary analytical method. MR-Egger and the weighted median (IVW) methods were utilized as supplementary analytical techniques. Sensitivity analyses including MR-Pleiotropy RESidual Sum and Outlier method (PRESSO), Cochran's Q test, and MR-Egger intercept test were conducted to address heterogeneity and horizontal pleiotropy. Results Ceramide (d42:2) and triacylglycerol (56:3) were found to be associated with an increased risk of ED, while phosphatidylethanolamine (O-18:1_18:2) and phosphatidylinositol (18:1_18:1) were linked to a decreased risk of ED. Interleukin-1α (IL-1α), IL-7, IL-17C, and the tumor necrosis factor (TNF) receptor superfamily member 9 (TNFRSF9) positively affected ED. Conversely, leukemia inhibitory factor and urokinase-type plasminogen activator (uPA) showed a negative impact. Mediation analysis indicated that the uPA mediated between Triacylglycerol (56:3) and ED, accounting for a mediation proportion of -14.71%. Conclusions There was a causal relationship between plasma lipidome and circulating inflammatory proteins with ED. Circulating inflammatory proteins appeared to mediate between triacylglycerol (56:3) levels and ED.
Collapse
Affiliation(s)
- Jiacheng Yu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peihe Liang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Chaparro V, Leroux LP, Lebourg A, Chagneau S, Graber TE, Alain T, Jaramillo M. Leukemia inhibitory factor drives transcriptional programs that promote lipid accumulation and M2 polarization in macrophages. J Leukoc Biol 2024; 117:qiae178. [PMID: 39178293 DOI: 10.1093/jleuko/qiae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/24/2024] [Accepted: 08/22/2024] [Indexed: 08/25/2024] Open
Abstract
Leukemia inhibitory factor, a member of the interleukin-6 cytokine family, plays a central role in homeostasis and disease. Interestingly, some of the pleiotropic effects of leukemia inhibitory factor have been attributed to the modulation of macrophage functions although the molecular underpinnings have not been explored at a genome-wide scale. Herein, we investigated leukemia inhibitory factor-driven transcriptional changes in murine bone marrow-derived macrophages by RNA sequencing. In silico analyses revealed a selective and time-dependent remodeling of macrophage gene expression programs associated with lipid metabolism and cell activation. Accordingly, a subset of leukemia inhibitory factor-upregulated transcripts related to cholesterol metabolism and lipid internalization was validated by real-time quantitative polymerase chain reaction. This was accompanied by a leukemia inhibitory factor-enhanced capacity for lipid accumulation in macrophages upon incubation with oxidized low-density lipoprotein. Mechanistically, leukemia inhibitory factor triggered the phosphorylation (Y705 and S727) and nuclear translocation of the transcription factor STAT3 in bone marrow-derived macrophages. Consistent with this, ingenuity pathway analysis identified STAT3 as an upstream regulator of a subset of transcripts, including Il4ra, in leukemia inhibitory factor-treated macrophages. Notably, leukemia inhibitory factor priming enhanced bone marrow-derived macrophage responses to interleukin-4-mediated M2 polarization (i.e. increased arginase activity and accumulation of transcripts encoding for M2 markers). Conversely, leukemia inhibitory factor stimulation had no significant effect in bone marrow-derived macrophage responses to M1-polarizing stimuli (interferon-γ and lipopolysaccharide). Thus, our study provides insight into the transcriptional landscape of leukemia inhibitory factor-treated macrophages, shedding light on its role in lipid metabolism and M2 polarization responses. A better understanding of the regulatory mechanisms governing leukemia inhibitory factor-driven changes might help informing novel therapeutic approaches aiming to reprogram macrophage phenotypes in diseased states (e.g. cancer, atherosclerosis, and infection).
Collapse
Affiliation(s)
- Visnu Chaparro
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Louis-Philippe Leroux
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Aurore Lebourg
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Sophie Chagneau
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, 401 Smith Rd. Ottawa, ON K1H 8L1, Canada
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, 401 Smith Rd. Ottawa, ON K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, 75 Laurier Ave E. University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Maritza Jaramillo
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| |
Collapse
|
22
|
Liu R, Cui H, Li D, Guo X, Zhang Z, Tan S, Zhu X. Roles and Mechanisms of Ferroptosis in Sorafenib Resistance for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:2493-2504. [PMID: 39717509 PMCID: PMC11665174 DOI: 10.2147/jhc.s500084] [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: 10/10/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent malignant tumor, characterized by a poor prognosis. In recent decades, both the incidence and mortality rates of HCC have risen sharply. Sorafenib has emerged as the first conventional drug approved by the US Food and Drug Administration for first-line treatment in advanced HCC patients due to its favorable safety profile. However, its effectiveness is severely hindered by acquired drug resistance, which leads to only approximately 30% of HCC patients benefited from sorafenib therapy. Sorafenib resistance involves various mechanisms that inhibit cellular uptake of iron and reactive oxygen species (ROS). Consequently, ferroptosis a novel form of cell death contingent upon the accumulation of intracellular iron and ROS plays a critical role in mediating sorafenib resistance through the Hippo YAP pathway or Keap1-Nrf2 system. This review aimed to comprehensively elucidate the mechanisms underlying sorafenib resistance in HCC, particularly focusing on ferroptosis and its pathways, to provide valuable insights into targeting ferroptosis or its pathways for sorafenib-resistant HCC treatment.
Collapse
Affiliation(s)
- Ruyuan Liu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Huanyu Cui
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Di Li
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Xuefeng Guo
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Zhengbao Zhang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Shengkui Tan
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, People’s Republic of China
| | - Xiaonian Zhu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| |
Collapse
|
23
|
Cui YH, Wei J, Fan H, Li W, Zhao L, Wilkinson E, Peterson J, Xie L, Zou Z, Yang S, Applebaum MA, Kline J, Chen J, He C, He YY. Targeting DTX2/UFD1-mediated FTO degradation to regulate antitumor immunity. Proc Natl Acad Sci U S A 2024; 121:e2407910121. [PMID: 39661064 DOI: 10.1073/pnas.2407910121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Here, we show that vitamin E succinate (VES) acts as a degrader for the m6A RNA demethylase fat mass and obesity-associated protein (FTO), thus suppressing tumor growth and resistance to immunotherapy. FTO is ubiquitinated by its E3 ligase DTX2, followed by UFD1 recruitment and subsequent degradation in the proteasome. VES binds to FTO and DTX2, leading to enhanced FTO-DTX2 interaction, FTO ubiquitination, and degradation in FTO-dependent tumor cells. VES suppressed tumor growth and enhanced antitumor immunity and response to immunotherapy in vivo in mouse models. Genetic FTO knockdown or VES treatment increased m6A methylation in the LIF (Leukemia Inhibitory Factor) gene and decreased LIF mRNA decay, and thus sensitized melanoma cells to T cell-mediated cytotoxicity. Taken together, our findings reveal the underlying molecular mechanism for FTO protein degradation and identify a dietary degrader for FTO that inhibits tumor growth and overcomes immunotherapy resistance.
Collapse
Affiliation(s)
- Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Jiangbo Wei
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Hao Fan
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Wenlong Li
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Lijie Zhao
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637
| | - Jack Peterson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- The College, University of Chicago, Chicago, IL 60637
| | - Lishi Xie
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Zhongyu Zou
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Mark A Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637
| | - Justin Kline
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Jing Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Chuan He
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
- HHMI, University of Chicago, Chicago, IL 60637
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
24
|
Isola JVV, Biswas S, Jayarathne H, Hubbart CR, Hense JD, Matsuzaki S, Kinter MT, Humphries KM, Ocañas SR, Sadagurski M, Stout MB. Canagliflozin treatment prevents follicular exhaustion and attenuates hallmarks of ovarian aging in genetically heterogenous mice. GeroScience 2024:10.1007/s11357-024-01465-w. [PMID: 39672978 DOI: 10.1007/s11357-024-01465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian aging is characterized by declines in follicular reserve and the emergence of mitochondrial dysfunction, reactive oxygen species production, inflammation, and fibrosis, which eventually results in menopause. Menopause is associated with increased systemic aging and the development of numerous comorbidities; therefore, the attenuation of ovarian aging could also delay systemic aging processes in women. Recent work has established that the anti-diabetic drug Canagliflozin (Cana), a sodium-glucose transporter 2 inhibitor, elicits benefits on aging-related outcomes, likely through the modulation of nutrient-sensing pathways and metabolic homeostasis. Given that nutrient-sensing pathways play a critical role in controlling primordial follicle activation, we sought to determine if chronic Cana administration would delay ovarian aging and curtail the emergence of pathological hallmarks associated with reproductive senescence. We found that mice receiving Cana maintained their ovarian reserve through 12 months of age, which was associated with declines in primordial follicles FoxO3a phosphorylation, a marker of activation, when compared to the age-matched controls. Furthermore, Cana treatment led to decreased collagen, lipofuscin, and T cell accumulation at 12 months of age. Whole ovary transcriptomic and proteomic analyses revealed subtle improvements, predominantly in mitochondrial function and the regulation of cellular proliferation. Pathway analyses of the transcriptomic data revealed a downregulation in cell proliferation and mitochondrial dysfunction signatures, with an upregulation of oxidative phosphorylation. Pathway analyses of the proteomic data revealed declines in signatures associated with PI3K/AKT activity and lymphocyte accumulation. Collectively, we demonstrate that Cana treatment can delay ovarian aging in mice and could potentially have efficacy for delaying ovarian aging in women.
Collapse
Affiliation(s)
- José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Subhasri Biswas
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Hashan Jayarathne
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Chase R Hubbart
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Jessica D Hense
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
25
|
Akula S, Alvarado-Vazquez A, Haide Mendez Enriquez E, Bal G, Franke K, Wernersson S, Hallgren J, Pejler G, Babina M, Hellman L. Characterization of Freshly Isolated Human Peripheral Blood B Cells, Monocytes, CD4+ and CD8+ T Cells, and Skin Mast Cells by Quantitative Transcriptomics. Int J Mol Sci 2024; 25:13050. [PMID: 39684762 DOI: 10.3390/ijms252313050] [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: 11/12/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Quantitative transcriptomics offers a new way to obtain a detailed picture of freshly isolated cells. By direct isolation, the cells are unaffected by in vitro culture, and the isolation at cold temperatures maintains the cells relatively unaltered in phenotype by avoiding activation through receptor cross-linking or plastic adherence. Simultaneous analysis of several cell types provides the opportunity to obtain detailed pictures of transcriptomic differences between them. Here, we present such an analysis focusing on four human blood cell populations and compare those to isolated human skin mast cells. Pure CD19+ peripheral blood B cells, CD14+ monocytes, and CD4+ and CD8+ T cells were obtained by fluorescence-activated cell sorting, and KIT+ human connective tissue mast cells (MCs) were purified by MACS sorting from healthy skin. Detailed information concerning expression levels of the different granule proteases, protease inhibitors, Fc receptors, other receptors, transcription factors, cell signaling components, cytoskeletal proteins, and many other protein families relevant to the functions of these cells were obtained and comprehensively discussed. The MC granule proteases were found exclusively in the MC samples, and the T-cell granzymes in the T cells, of which several were present in both CD4+ and CD8+ T cells. High levels of CD4 were also observed in MCs and monocytes. We found a large variation between the different cell populations in the expression of Fc receptors, as well as for lipid mediators, proteoglycan synthesis enzymes, cytokines, cytokine receptors, and transcription factors. This detailed quantitative comparative analysis of more than 780 proteins of importance for the function of these populations can now serve as a good reference material for research into how these entities shape the role of these cells in immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Abigail Alvarado-Vazquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Erika Haide Mendez Enriquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Sara Wernersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
26
|
Zhang Y, Cong Y, Du J, Guo D, Huang J, Pan J, Liang Y, Zhang J, Ye Z, Liu Y, Zhou Y. Lif-deficiency promote systemic Iron metabolism disorders and increases the susceptibility of osteoblasts to ferroptosis. Bone 2024; 189:117266. [PMID: 39341481 DOI: 10.1016/j.bone.2024.117266] [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: 07/29/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Leukemia inhibitory factor (LIF) is a multifunctional cytokine that plays a crucial role in various biological processes. However, LIF involvement in iron metabolism remains almost unexplored. This study aimed to explore the impact of LIF on systemic iron transportation and its potential role in ferroptosis in osteoblasts. We observed that the Lif-deficient (Lif-/-) mice is characterized by a reduction in bone mass and a decrease in bone mineral density compared with wild-type (WT) mice. Energy-dispersive X-ray spectroscopy revealed a marked increase in iron content on the surface of femurs from Lif-/- mice. Meanwhile, iron stores test lower iron levels in the spleens and higher levels in the femurs of Lif-/- mice. Besides, Lif-/- mice display increased levels of serum iron, total iron-binding capacity, unsaturated iron-binding capacity, and transferrin saturation and serum ferritin relative to WT mice. Hepcidin mRNA expression reduction in the liver of Lif-/- mice. It also holds true in the AML-12 hepatocyte cell line after Lif-knockdown. Immunohistochemistry and RT-PCR revealed elevated ferroportin (FPN) in duodenal cells of Lif-/- mice. Lif-deficiency decreases SLC7A11 levels in osteoblasts. In addition, overexpression of LIF downregulates CD71, DCYTB, and DMT1, thereby reducing iron uptake in iron-overloaded cells. Femur immunohistochemistry (IHC) revealed increased ACSL4 and decreased GPX4 and SLC7A11, indicating an increase in ferroptosis of osteoblasts in Lif-/- mice. Whole-transcriptome sequencing showed gene expression changes after Lif-knockdown, exhibiting a negative correlation with genes involved in long-chain fatty acid transport, mitochondrial organization, and the p38 MAPK signaling pathway. These results demonstrate that Lif-deficiency alter systemic iron metabolism and increases the susceptibility of osteoblasts to ferroptosis.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yaqi Cong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Juan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Donghua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jing Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Junchen Pan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Youde Liang
- The People's Hospital of Baoan Shenzhen, Shenzhen, China
| | - Jiali Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Yi Liu
- Department of Stomatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi 435002, China.
| | - Yi Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Center for Prosthodontics and Implant Dentistry, Optics Valley Branch, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
27
|
Wiens KR, Wasti N, Ulloa OO, Klegeris A. Diversity of Microglia-Derived Molecules with Neurotrophic Properties That Support Neurons in the Central Nervous System and Other Tissues. Molecules 2024; 29:5525. [PMID: 39683685 DOI: 10.3390/molecules29235525] [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: 10/02/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain immune cells, support neurons by producing several established neurotrophic molecules including glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Modern analytical techniques have identified numerous phenotypic states of microglia, each associated with the secretion of a diverse set of substances, which likely include not only canonical neurotrophic factors but also other less-studied molecules that can interact with neurons and provide trophic support. In this review, we consider the following eight such candidate cytokines: oncostatin M (OSM), leukemia inhibitory factor (LIF), activin A, colony-stimulating factor (CSF)-1, interleukin (IL)-34, growth/differentiation factor (GDF)-15, fibroblast growth factor (FGF)-2, and insulin-like growth factor (IGF)-2. The available literature provides sufficient evidence demonstrating murine cells produce these cytokines and that they exhibit neurotrophic activity in at least one neuronal model. Several distinct types of neurotrophic activity are identified that only partially overlap among the cytokines considered, reflecting either their distinct intrinsic properties or lack of comprehensive studies covering the full spectrum of neurotrophic effects. The scarcity of human-specific studies is another significant knowledge gap revealed by this review. Further studies on these potential microglia-derived neurotrophic factors are warranted since they may be used as targeted treatments for diverse neurological disorders.
Collapse
Affiliation(s)
- Kennedy R Wiens
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Naved Wasti
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Omar Orlando Ulloa
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
28
|
Salmasi S, Heidar MS, Khaksary Mahabady M, Rashidi B, Mirzaei H. MicroRNAs, endometrial receptivity and molecular pathways. Reprod Biol Endocrinol 2024; 22:139. [PMID: 39529197 PMCID: PMC11552404 DOI: 10.1186/s12958-024-01304-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
MicroRNAs (miRNAs) are a type of specific molecules that control the activities of the uterus, such as the process of cellular maturing and evolution. A lot of substances like growth factors, cytokines, and transcription factors play a role in embryo-endometrial interaction. MiRNAs could regulate various these factors by attaching to the 3' UTR of their mRNAs. Moreover, current research show that miRNAs participate in formation of blood vessels in endometrium (miR-206, miR-17-5p, miR-16-5p…), decidualization (miR-154, miR-181, miR-9…), epithelial-mesenchymal transition (miR-30a-3p), immune response (miR-888, miR-376a, miR-300…) embryo attachment (miR-145, miR-27a,451…) and pinopod formation (mir-223-3p, mir-449a, mir-200c). In this study, the focus is on the role of miRNAs in managing the uterus' receptivity to an embryo and its ability to facilitate attachment. More specifically, we are exploring the mechanisms by which miRNAs regulate the presence of specific molecules involved in this crucial physiological process.
Collapse
Affiliation(s)
- Soheila Salmasi
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Saeed Heidar
- Faculty of life sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
29
|
Ebert ET, Schwinghamer KM, Siahaan TJ. Delivery of Neuroregenerative Proteins to the Brain for Treatments of Neurodegenerative Brain Diseases. Life (Basel) 2024; 14:1456. [PMID: 39598254 PMCID: PMC11595909 DOI: 10.3390/life14111456] [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: 09/21/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Neurodegenerative brain diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), and Parkinson's disease (PD) are difficult to treat. Unfortunately, many therapeutic agents for neurodegenerative disease only halt the progression of these diseases and do not reverse neuronal damage. There is a demand for finding solutions to reverse neuronal damage in the central nervous system (CNS) of patients with neurodegenerative brain diseases. Therefore, the purpose of this review is to discuss the potential for therapeutic agents like specific neurotrophic and growth factors in promoting CNS neuroregeneration in brain diseases. We discuss how BDNF, NGF, IGF-1, and LIF could potentially be used for the treatment of brain diseases. The molecule's different mechanisms of action in stimulating neuroregeneration and methods to analyze their efficacy are described. Methods that can be utilized to deliver these proteins to the brain are also discussed.
Collapse
Affiliation(s)
| | | | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; (E.T.E.); (K.M.S.)
| |
Collapse
|
30
|
Ou Y, Fan L, Wang X, Xia H, Cheng M, Huang J, Liang Y, Wang Y, Zhou Y. Leukemia inhibitory factor protects against experimental periodontitis through immuno-modulations of both macrophages and periodontal ligament fibroblasts. J Periodontol 2024; 95:1073-1085. [PMID: 38488753 DOI: 10.1002/jper.23-0607] [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: 10/24/2023] [Revised: 01/08/2024] [Accepted: 02/15/2024] [Indexed: 12/05/2024]
Abstract
BACKGROUND To explore the role of leukemia inhibitory factor (LIF) in periodontitis via in vivo and in vitro experiments. METHODS The second upper molar of LIF knockout mice and their wild-type littermates were ligated for 8 days. Micro-computed tomography (micro-CT), histological analysis, and quantitative real-time polymerase chain reaction (qRT-PCR) were performed. The expression levels of proinflammatory cytokines were examined in mouse bone marrow derived macrophages and human periodontal ligament fibroblasts (HPDLFs) after lipopolysaccharide (LPS) treatment. RESULTS LIF deficiency promoted alveolar bone loss, inflammatory cells infiltration, osteoclasts formation and collagen fiber degradation in ligature-induced mouse, along with higher expressions of proinflammatory cytokines, including interleukin-6 (IL6), IL-1β (IL1B), tumor necrosis factor-α (TNFA), matrix metalloproteinase 13 (MMP13), and RANKL/OPG ratio. Additionally, LIF deletion led to higher expression levels of these proinflammatory cytokines in mouse bone marrow-derived macrophages from both femur and alveolar bone and HPDLFs when treated with LPS. Administration of recombined LIF attenuated TNFA, IL1B, and RANKL/OPG ratio in HPDLFs. CONCLUSIONS These findings indicate that LIF deficiency promotes the progress of periodontitis via modulating immuno-inflammatory responses of macrophages and periodontal ligament fibroblasts, and the application of LIF may be an adjunctive treatment for periodontitis to resolute inflammation.
Collapse
Affiliation(s)
- Yanjing Ou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
- Postdoctoral Workstation & Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, PR China
| | - Le Fan
- Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Xiaoqi Wang
- Department of Stomatology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
| | - Mengwen Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
| | - Jing Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
| | - Youde Liang
- Department of Stomatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, Guangdong, PR China
| | - Yining Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
| | - Yi Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|
31
|
Randolph L, Joshi J, Rodriguez Sanchez AL, Pratap UP, Gopalam R, Chen Y, Lai Z, Santhamma B, Kost ER, Nair HB, Vadlamudi RK, Subbarayalu P, Viswanadhapalli S. Significance of LIF/LIFR Signaling in the Progression of Obesity-Driven Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3630. [PMID: 39518071 PMCID: PMC11545110 DOI: 10.3390/cancers16213630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/20/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
American women with obesity have an increased incidence of triple-negative breast cancer (TNBC). The impact of obesity conditions on the tumor microenvironment is suspected to accelerate TNBC progression; however, the specific mechanism(s) remains elusive. This study explores the hypothesis that obesity upregulates leukemia inhibitory factor receptor (LIFR) oncogenic signaling in TNBC and assesses the efficacy of LIFR inhibition with EC359 in blocking TNBC progression. TNBC cell lines were co-cultured with human primary adipocytes, or adipocyte-conditioned medium, and treated with EC359. The effects of adiposity were measured using cell viability, colony formation, and invasion assays. Mechanistic studies utilized RNA-Seq, Western blotting, RT-qPCR, and reporter gene assays. The therapeutic potential of EC359 was tested using xenograft and patient-derived organoid (PDO) models. The results showed that adipose conditions increased TNBC cell proliferation and invasion, and these effects correlated with enhanced LIFR signaling. Accordingly, EC359 treatment reduced cell viability, colony formation, and invasion under adipose conditions and blocked adipose-mediated organoid growth and TNBC xenograft tumor growth. RNA-Seq analysis identified critical pathways modulated by LIF/LIFR signaling in diet-induced obesity mouse models. These findings suggest that adiposity contributes to TNBC progression via the activation of the LIF/LIFR pathway, and LIFR inhibition with EC359 represents a promising therapeutic approach for obesity-associated TNBC.
Collapse
Affiliation(s)
- Lois Randolph
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
| | - Jaitri Joshi
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Alondra Lee Rodriguez Sanchez
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
| | - Rahul Gopalam
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.C.); (Z.L.)
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.C.); (Z.L.)
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | | | - Edward R. Kost
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
| | | | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Panneerdoss Subbarayalu
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.C.); (Z.L.)
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (L.R.); (A.L.R.S.); (U.P.P.); (R.G.); (E.R.K.); (R.K.V.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
32
|
Rapacciuolo P, Finamore C, Giorgio CD, Fiorillo B, Massa C, Urbani G, Marchianò S, Bordoni M, Cassiano C, Morretta E, Spinelli L, Lupia A, Moraca F, Biagioli M, Sepe V, Monti MC, Catalanotti B, Fiorucci S, Zampella A. Design, Synthesis, and Pharmacological Evaluation of Dual FXR-LIFR Modulators for the Treatment of Liver Fibrosis. J Med Chem 2024; 67:18334-18355. [PMID: 39382988 DOI: 10.1021/acs.jmedchem.4c01651] [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: 10/11/2024]
Abstract
Although multiple approaches have been suggested, treating mild-to-severe fibrosis in the context of metabolic dysfunction associated with liver disease (MASLD) remains a challenging area in drug discovery. Pathogenesis of liver fibrosis is multifactorial, and pathogenic mechanisms are deeply intertwined; thus, it is well accepted that future treatment requires the development of multitarget modulators. Harnessing the 3,4,5-trisubstituted isoxazole scaffold, previously described as a key moiety in Farnesoid X receptor (FXR) agonism, herein we report the discovery of a novel class of hybrid molecules endowed with dual activity toward FXR and the leukemia inhibitory factor receptor (LIFR). Up to 27 new derivatives were designed and synthesized. The pharmacological characterization of this series resulted in the identification of 3a as a potent FXR agonist and LIFR antagonist with excellent ADME properties. In vitro and in vivo characterization identified compound 3a as the first-in-class hybrid LIFR inhibitor and FXR agonist that protects against the development of acute liver fibrosis and inflammation.
Collapse
Affiliation(s)
- Pasquale Rapacciuolo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Claudia Finamore
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Cristina Di Giorgio
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Carmen Massa
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Silvia Marchianò
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Martina Bordoni
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Chiara Cassiano
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Elva Morretta
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Lucio Spinelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Antonio Lupia
- Department of Life and Environmental Sciences, University of Cagliari, Via Università, 40, Cagliari 09124, Italy
| | - Federica Moraca
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Maria Chiara Monti
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Bruno Catalanotti
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| |
Collapse
|
33
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [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: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
34
|
Scott EN, Ye C, Yano H, Lipatova Z, Brunazzi E, Vignali KM, Workman CJ, Vignali DA. Ebi3 Binding to IFN-γ and IL-10 Limits Their Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1115-1124. [PMID: 39240167 PMCID: PMC11458358 DOI: 10.4049/jimmunol.2400236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
EBV-induced gene 3 (Ebi3) is a β subunit within the IL-12 cytokine family that canonically binds to α subunits p19, p28, or p35 to form the heterodimeric cytokines IL-39, IL-27, and IL-35, respectively. In the last decade, the binding partners for Ebi3 have continued to expand to include IL-6 and the other IL-12 family β subunit p40, revealing the possibility that Ebi3 may be able to bind to other cytokines and have distinct functions. We first explored this possibility utilizing an in vivo mouse model of regulatory T cell-restricted deletions of the subunits composing the cytokine IL-35, p35, and Ebi3, and we observed a differential impact on CD8+ T cell inhibitory receptor expression despite comparable reduction in tumor growth. We then screened the ability of Ebi3 to bind to different cytokines with varying structural resemblance to the IL-12 family α subunits. These in vitro screens revealed extracellular binding of Ebi3 to both IFN-γ and IL-10. Ebi3 bound to IFN-γ and IL-10 abrogated signal transduction and downstream functions of both cytokines. Lastly, we validated that extracellular complex formation after mixing native proteins resulted in loss of function. These data suggest that secreted partnerless Ebi3 may bind to cytokines within the extracellular microenvironment and act as a cytokine sink, further expanding the potential immunological impact of Ebi3.
Collapse
Affiliation(s)
- Ellen N. Scott
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Cheng Ye
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Present address: Neurophth Therapeutics, Minhang District, Shanghai, China
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Present address and affiliation: Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY
| | - Zhanna Lipatova
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Erin Brunazzi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Kate M. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Dario A.A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| |
Collapse
|
35
|
Alesutan I, Razazian M, Luong TTD, Estepa M, Pitigala L, Henze LA, Obereigner J, Mitter G, Zickler D, Schuchardt M, Deisl C, Makridakis M, Gollmann-Tepeköylü C, Pasch A, Cejka D, Suessner S, Antlanger M, Bielesz B, Müller M, Vlahou A, Holfeld J, Eckardt KU, Voelkl J. Augmentative effects of leukemia inhibitory factor reveal a critical role for TYK2 signaling in vascular calcification. Kidney Int 2024; 106:611-624. [PMID: 39084258 DOI: 10.1016/j.kint.2024.07.011] [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: 08/04/2023] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
Medial vascular calcification in chronic kidney disease (CKD) involves pro-inflammatory pathways induced by hyperphosphatemia. Several interleukin 6 family members have been associated with pro-calcific effects in vascular smooth muscle cells (VSMCs) and are considered as therapeutic targets. Therefore, we investigated the role of leukemia inhibitory factor (LIF) during VSMC calcification. LIF expression was found to be increased following phosphate exposure of VSMCs. LIF supplementation aggravated, while silencing of endogenous LIF or LIF receptor (LIFR) ameliorated the pro-calcific effects of phosphate in VSMCs. The soluble LIFR mediated antagonistic effects towards LIF and reduced VSMC calcification. Mechanistically, LIF induced phosphorylation of the non-receptor tyrosine-protein kinase 2 (TYK2) and signal transducer and activator of transcription-3 (STAT3) in VSMCs. TYK2 inhibition by deucravacitinib, a selective, allosteric oral immunosuppressant used in psoriasis treatment, not only blunted the effects of LIF, but also interfered with the pro-calcific effects induced by phosphate. Conversely, TYK2 overexpression aggravated VSMC calcification. Ex vivo calcification of mouse aortic rings was ameliorated by Tyk2 pharmacological inhibition and genetic deficiency. Cholecalciferol-induced vascular calcification in mice was improved by Tyk2 inhibition and in the Tyk2-deficient mice. Similarly, calcification was ameliorated in Abcc6/Tyk2-deficient mice after adenine/high phosphorus-induced CKD. Thus, our observations indicate a role for LIF in CKD-associated vascular calcification. Hence, the effects of LIF identify a central pro-calcific role of TYK2 signaling, which may be a future target to reduce the burden of vascular calcification in CKD.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Cells, Cultured
- Disease Models, Animal
- Leukemia Inhibitory Factor/metabolism
- Leukemia Inhibitory Factor/genetics
- Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism
- Leukemia Inhibitory Factor Receptor alpha Subunit/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phosphates/metabolism
- Phosphorylation
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- STAT3 Transcription Factor/metabolism
- TYK2 Kinase/metabolism
- TYK2 Kinase/genetics
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/etiology
- Vascular Calcification/genetics
Collapse
Affiliation(s)
- Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Mehdi Razazian
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Trang T D Luong
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Lakmi Pitigala
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Laura A Henze
- Department of Internal Medicine and Cardiology, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Jakob Obereigner
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Gregor Mitter
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany; Faculty of Medicine, Medical School Berlin, Berlin, Germany
| | - Christine Deisl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Manousos Makridakis
- Center of Systems Biology, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | | | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria; Calciscon AG, Biel, Switzerland
| | - Daniel Cejka
- Internal Medicine III-Nephrology, Transplantation Medicine, Rheumatology, Ordensklinikum Linz, Linz, Austria
| | | | - Marlies Antlanger
- Department of Internal Medicine 2, Kepler University Hospital and Johannes Kepler University, Linz, Austria
| | - Bernhard Bielesz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Antonia Vlahou
- Center of Systems Biology, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Johannes Holfeld
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria; Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
36
|
Li J, Wang Y, Wang Z, Wei Y, Diao P, Wu Y, Wang D, Jiang H, Wang Y, Cheng J. Super-Enhancer Driven LIF/LIFR-STAT3-SOX2 Regulatory Feedback Loop Promotes Cancer Stemness in Head and Neck Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404476. [PMID: 39206755 PMCID: PMC11516160 DOI: 10.1002/advs.202404476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Super-enhancers (SEs) have been recognized as key epigenetic regulators underlying cancer stemness and malignant traits by aberrant transcriptional control and promising therapeutic targets against human cancers. However, the SE landscape and their roles during head and neck squamous cell carcinoma (HNSCC) development especially in cancer stem cells (CSCs) maintenance remain underexplored yet. Here, we identify leukemia inhibitory factor (LIF)-SE as a representative oncogenic SE to activate LIF transcription in HNSCC. LIF secreted from cancer cells and cancer-associated fibroblasts promotes cancer stemness by driving SOX2 transcription in an autocrine/paracrine manner, respectively. Mechanistically, enhancer elements E1, 2, 4 within LIF-SE recruit SOX2/SMAD3/BRD4/EP300 to facilitate LIF transcription; LIF activates downstream LIFR-STAT3 signaling to drive SOX2 transcription, thus forming a previously unknown regulatory feedback loop (LIF-SE-LIF/LIFR-STAT3-SOX2) to maintain LIF overexpression and CSCs stemness. Clinically, increased LIF abundance in clinical samples correlate with malignant clinicopathological features and patient prognosis; higher LIF concentrations in presurgical plasma dramatically diminish following cancer eradication. Therapeutically, pharmacological targeting LIF-SE-LIF/LIFR-STAT3 significantly impairs tumor growth and reduces CSC subpopulations in xenograft and PDX models. Our findings reveal a hitherto uncharacterized LIF-SE-mediated auto-regulatory loop in regulating HNSCC stemness and highlight LIF as a novel noninvasive biomarker and potential therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Jin Li
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yuhan Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Ziyu Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yuxiang Wei
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Pengfei Diao
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yaping Wu
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Dongmiao Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
| | - Hongbing Jiang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yanling Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
| | - Jie Cheng
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| |
Collapse
|
37
|
Zhu M, Yu R, Liu Y, Geng X, Liu Q, Liu S, Zhu Y, Li G, Guo Y, Xi X, Du B. LncRNA H19 Participates in Leukemia Inhibitory Factor Mediated Stemness Promotion in Colorectal Cancer Cells. Biochem Genet 2024; 62:3695-3708. [PMID: 38198021 DOI: 10.1007/s10528-023-10627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024]
Abstract
Colorectal cancer (CRC) is a common human malignancy and the third leading cause of cancer-related death worldwide. Cancer stem cells (CSCs) were considered to play important roles in the genesis and development of many tumors. In recent years, it has been observed that leukemia inhibitory factor (LIF) might be involved in the regulation of stemness in cancer cells. In this study, we observed that LIF could increase the spheroid formation and stemness marker expression (inculding Nanog and SOX2) in CRC cell lines, such as HCT116 and Caco2 cells. Meanwhile, we also observed that LIF could upregulate LncRNA H19 expression via PI3K/AKT pathway. Knockdown of the expression of LncRNA H19 could decrease the spheroid formation and SOX2 expression in LIF-treated HCT116 and Caco2 cells, and thereby LncRNA H19 knockdown could compensate for the stemness enhancement effects induced by LIF. Our results indicated that LncRNA H19 might participate in the stemness promotion of LIF in CRC cells.
Collapse
Affiliation(s)
- Min Zhu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ruihong Yu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Yirui Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Xiaoqing Geng
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Qiong Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Shuaitong Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Yunhe Zhu
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Gang Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yang Guo
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Xueyan Xi
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China.
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Boyu Du
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China.
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
38
|
Seifati SM, Zare F, Bafghi SAM, Hadinedoushan H. Impact of anti leukemia inhibitory factor antibody on immune related gene expression in breast cancer Balb/c mouse model. Sci Rep 2024; 14:20403. [PMID: 39223212 PMCID: PMC11369080 DOI: 10.1038/s41598-024-71014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Leukemia inhibitory factor (LIF) is involved in the progression of different cancers. In this study, we investigated the effect of anti-LIF antibodies on immune-related gene expression in the Balb/c mouse model of breast cancer. To immunize mice against LIF, recombinant LIF with Freund adjuvant was injected into the test group, whereas the control group received phosphate-buffered saline with adjuvant. Tumor induction (4T1 cell line) was performed by increasing the antibody titer. The expression of immune-related genes was evaluated by real-time PCR. The anti-LIF titer was significantly increased in the immunized group. The expression of genes related to the differentiation of T helper (Th)-1, Th-2, and Th-17 cells was significantly higher in the immunized group than in the control group. In addition, anti-LIF did not have a significant effect on the expression of genes related to the differentiation of regulatory T cells, and immune checkpoint-associated genes. Additionally, the test group had higher survival and lower tumor development rates. The results demonstrated that the anti-LIF antibody may potentially play a role in the differentiation of immune cells or immune responses. However, further studies utilizing advanced techniques are necessary to validate its function.
Collapse
Affiliation(s)
- Seyed Mohammad Seifati
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
| | | | - Hossein Hadinedoushan
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran.
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
39
|
Agca S, Kir S. The role of interleukin-6 family cytokines in cancer cachexia. FEBS J 2024; 291:4009-4023. [PMID: 38975832 DOI: 10.1111/febs.17224] [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: 11/30/2023] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024]
Abstract
Cachexia is a wasting syndrome that manifests in more than half of all cancer patients. Cancer-associated cachexia negatively influences the survival of patients and their quality of life. It is characterized by a rapid loss of adipose and skeletal muscle tissues, which is partly mediated by inflammatory cytokines. Here, we explored the crucial roles of interleukin-6 (IL-6) family cytokines, including IL-6, leukemia inhibitory factor, and oncostatin M, in the development of cancer cachexia. These cytokines have been shown to exacerbate cachexia by promoting the wasting of adipose and muscle tissues, activating mechanisms that enhance lipolysis and proteolysis. Overlapping effects of the IL-6 family cytokines depend on janus kinase/signal transducer and activator of transcription 3 signaling. We argue that the blockade of these cytokine pathways individually may fail due to redundancy and future therapeutic approaches should target common downstream elements to yield effective clinical outcomes.
Collapse
Affiliation(s)
- Samet Agca
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Serkan Kir
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| |
Collapse
|
40
|
Zhao S, Deng H, Lu Y, Tao Y, Li D, Jiang X, Wei X, Chen X, Ma F, Wang Y, Gou L, Yang J. Antagonist anti-LIF antibody derived from naive human scFv phage library inhibited tumor growth in mice. BMC Immunol 2024; 25:56. [PMID: 39169307 PMCID: PMC11340043 DOI: 10.1186/s12865-024-00636-w] [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: 05/07/2024] [Accepted: 06/25/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Leukemia inhibitory factor (LIF) is a multifunctional member of the IL-6 cytokine family that activates downstream signaling pathways by binding to the heterodimer consisting of LIFR and gp130 on the cell surface. Previous research has shown that LIF is highly expressed in various tumor tissues (e.g. pancreatic cancer, breast cancer, prostate cancer, and colorectal cancer) and promotes cancer cell proliferation, migration, invasion, and differentiation. Moreover, the overexpression of LIF correlates with poor clinicopathological characteristics. Therefore, we hypothesized that LIF could be a promising target for the treatment of cancer. In this work, we developed the antagonist antibody 1G11 against LIF and investigated its anti-tumor mechanism and its therapeutic efficacy in mouse models. RESULTS A series of single-chain variable fragments (scFvs) targeting LIF were screened from a naive human scFv phage library. These scFvs were reconstructed in complete IgG form and produced by the mammalian transient expression system. Among the antibodies, 1G11 exhibited the excellent binding activity to human, cynomolgus monkey and mouse LIF. Functional analysis demonstrated 1G11 could block LIF binding to LIFR and inhibit the intracellular STAT3 phosphorylation signal. Interestingly, 1G11 did not block LIF binding to gp130, another LIF receptor that is involved in forming the receptor complex together with LIFR. In vivo, intraperitoneal administration of 1G11 inhibited tumor growth in CT26 and MC38 models of colorectal cancer. IHC analysis demonstrated that p-STAT3 and Ki67 were decreased in tumor tissue, while c-caspase 3 was increased. Furthermore, 1G11 treatment improves CD3+, CD4 + and CD8 + T cell infiltration in tumor tissue. CONCLUSIONS We developed antagonist antibodies targeting LIF/LIFR signaling pathway from a naive human scFv phage library. Antagonist anti-LIF antibody exerts antitumor effects by specifically reducing p-STAT3. Further studies revealed that anti-LIF antibody 1G11 increased immune cell infiltration in tumor tissues.
Collapse
Grants
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-5-075 the CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
- 2018ZX09201018, 2017ZX09302010 the National Major Scientifc and Technological Special Project for Signifcant New Drugs Development
Collapse
Affiliation(s)
- Shengyan Zhao
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Han Deng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Lu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yiran Tao
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - David Li
- 503 Central Avenue, Sunnyvale, 94086, California, United States of America
| | - Xiaohua Jiang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xian Wei
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaofeng Chen
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanxin Ma
- Sound Biopharmaceuticals Co., Ltd, Tianfu International Bio-Town, Huigu Dong 2nd Road 8, 610200, Chengdu, Sichuan, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lantu Gou
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jinliang Yang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Chengdu, China.
| |
Collapse
|
41
|
Xie H, Jiang Y, Xiang Y, Wu B, Zhao J, Huang R, Wang M, Wang Y, Liu J, Wu D, Tian D, Bian E. Super-enhancer-driven LIF promotes the mesenchymal transition in glioblastoma by activating ITGB2 signaling feedback in microglia. Neuro Oncol 2024; 26:1438-1452. [PMID: 38554116 PMCID: PMC11300025 DOI: 10.1093/neuonc/noae065] [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: 09/12/2023] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND The mesenchymal (MES) subtype of glioblastoma (GBM) is believed to be influenced by both cancer cell-intrinsic alterations and extrinsic cellular interactions, yet the underlying mechanisms remain unexplored. METHODS Identification of microglial heterogeneity by bioinformatics analysis. Transwell migration, invasion assays, and tumor models were used to determine gene function and the role of small molecule inhibitors. RNA sequencing, chromatin immunoprecipitation, and dual-luciferase reporter assays were performed to explore the underlying regulatory mechanisms. RESULTS We identified the inflammatory microglial subtype of tumor-associated microglia (TAM) and found that its specific gene integrin beta 2 (ITGB2) was highly expressed in TAM of MES GBM tissues. Mechanistically, the activation of ITGB2 in microglia promoted the interaction between the SH2 domain of STAT3 and the cytoplasmic domain of ITGB2, thereby stimulating the JAK1/STAT3/IL-6 signaling feedback to promote the MES transition of GBM cells. Additionally, microglia communicated with GBM cells through the interaction between the receptor ITGB2 on microglia and the ligand ICAM-1 on GBM cells, while an increased secretion of ICAM-1 was induced by the proinflammatory cytokine leukemia inhibitory factor (LIF). Further studies demonstrated that inhibition of cyclin-dependent kinase 7 substantially reduced the recruitment of SNW1 to the super-enhancer of LIF, resulting in transcriptional inhibition of LIF. We identified notoginsenoside R1 as a novel LIF inhibitor that exhibited synergistic effects in combination with temozolomide. CONCLUSIONS Our research reveals that the epigenetic-mediated interaction of GBM cells with TAM drives the MES transition of GBM and provides a novel therapeutic avenue for patients with MES GBM.
Collapse
Affiliation(s)
- Han Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyi Jiang
- Institute of Health and Medical Technology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Yufei Xiang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Baoming Wu
- School of pharmacy, Anhui Medical University, Hefei, China
| | - Jiajia Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruixiang Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengting Wang
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunlong Wang
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Liu
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erbao Bian
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
Brown JC, Spielmann G, Yang S, Compton SLE, Jones LW, Irwin ML, Ligibel JA, Meyerhardt JA. Effects of exercise or metformin on myokine concentrations in patients with breast and colorectal cancer: A phase II multi-centre factorial randomized trial. J Cachexia Sarcopenia Muscle 2024; 15:1520-1527. [PMID: 38887915 PMCID: PMC11294014 DOI: 10.1002/jcsm.13509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Physical activity and metformin pharmacotherapy are associated with improved clinical outcomes in breast and colorectal cancer survivors. Myokines are cytokines secreted from skeletal muscle that may mediate these associations. METHODS This hypothesis-generating analysis used biospecimens collected from a multi-centre 2 × 2 factorial randomized design of 116 patients with stage I-III breast and colorectal cancer who were randomized to 12 weeks of (1) aerobic exercise (moderate intensity titrated to 220 min/week); (2) metformin (850 mg daily for 2 weeks and then titrated to 850 mg twice per day); (3) aerobic exercise and metformin; or (4) control. Fourteen myokines were quantified using a multiplex panel. Myokine concentrations were log-transformed, and main effects analyses were conducted using linear mixed-effects regression models. The type I error rate was controlled with the Holm sequential testing procedure. RESULTS Randomization to exercise increased leukaemia inhibitory factor (1.26 pg/mL, 95% confidence interval [CI]: 0.69, 1.84; adjusted P = 0.001) and interleukin-15 (2.23 pg/mL, 95% CI: 0.87, 3.60; adjusted P = 0.013) compared with randomization to no exercise. Randomization to metformin decreased apelin (-2.69 pg/mL, 95% CI: -4.31, -1.07; adjusted P = 0.014) and interleukin-15 (-1.74 pg/mL, 95% CI: -2.79, -0.69; adjusted P = 0.013) compared with randomization to no metformin. Metformin decreased myostatin, irisin, oncostatin M, fibroblast growth factor 21 and osteocrin; however, these changes were not statistically significant after correction for multiple comparisons. CONCLUSIONS This pilot study demonstrates that randomization to exercise and metformin elicit unique effects on myokine concentrations in cancer patients. This hypothesis-generating observation warrants further basic, translational and clinical investigation and replication.
Collapse
Affiliation(s)
- Justin C. Brown
- Pennington Biomedical Research CenterBaton RougeLAUSA
- LSU Health Sciences Center New Orleans School of MedicineNew OrleansLAUSA
- Stanley S. Scott Cancer CenterLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | | | | | | | - Lee W. Jones
- Memorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | | | | | | |
Collapse
|
43
|
Wu HM, Chen LH, Chiu WJ, Tsai CL. LIF-STAT signaling in decidual cells: a possible role in embryo implantation and early pregnancy. J Mol Endocrinol 2024; 73:e240006. [PMID: 38722222 PMCID: PMC11227039 DOI: 10.1530/jme-24-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/09/2024] [Indexed: 06/30/2024]
Abstract
In this study, we investigate the effects of miRNA-138-5p and probable G-protein coupled receptor 124 (GPR124)-regulated inflammasome and downstream leukemia inhibitory factor (LIF)-STAT and adhesion molecule signaling in human decidual stromal cells. After informed consent was obtained from women aged 25-38 years undergoing surgical termination of the normal pregnancy and spontaneous miscarriage after 6-9 weeks of gestation, human decidual stromal cells were extracted from the decidual tissue. Extracellular vesicles (EVs) with microRNA (miRNA) between cells have been regarded as critical factors for embryo-maternal interactions on embryo implantation and programming of human pregnancy. MicroRNA-138-5p acts as the transcriptional regulator of GPR124 and the mediator of downstream inflammasome. LIF-regulated STAT activation and expression of integrins might influence embryo implantation. Hence, a better understanding of LIF-STAT and adhesion molecule signaling would elucidate the mechanism of microRNA-138-5p- and GPR124-regulated inflammasome activation on embryo implantation and pregnancy. Our results show that microRNA-138-5p, purified from the EVs of decidual stromal cells, inhibits the expression of GPR124 and the inflammasome, and activates the expression of LIF-STAT and adhesion molecules in human decidual stromal cells. Additionally, the knockdown of GPR124 and NLRP3 through siRNA increases the expression of LIF-STAT and adhesion molecules. The findings of this study help us gain a better understanding the role of EVs, microRNA-138-5p, GPR124, inflammasomes, LIF-STAT, and adhesion molecules in embryo implantation and programming of human pregnancy.
Collapse
Affiliation(s)
- Hsien-Ming Wu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Liang-Hsuan Chen
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Wei-Jung Chiu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chia-Lung Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| |
Collapse
|
44
|
Gogoi M, Clark PA, Ferreira ACF, Rodriguez Rodriguez N, Heycock M, Ko M, Murphy JE, Chen V, Luan SL, Jolin HE, McKenzie ANJ. ILC2-derived LIF licences progress from tissue to systemic immunity. Nature 2024; 632:885-892. [PMID: 39112698 PMCID: PMC11338826 DOI: 10.1038/s41586-024-07746-w] [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: 09/18/2023] [Accepted: 06/24/2024] [Indexed: 08/17/2024]
Abstract
Migration and homing of immune cells are critical for immune surveillance. Trafficking is mediated by combinations of adhesion and chemokine receptors that guide immune cells, in response to chemokine signals, to specific locations within tissues and the lymphatic system to support tissue-localized immune reactions and systemic immunity1,2. Here we show that disruption of leukaemia inhibitory factor (LIF) production from group 2 innate lymphoid cells (ILC2s) prevents immune cells leaving the lungs to migrate to the lymph nodes (LNs). In the absence of LIF, viral infection leads to plasmacytoid dendritic cells (pDCs) becoming retained in the lungs where they improve tissue-localized, antiviral immunity, whereas chronic pulmonary allergen challenge leads to marked immune cell accumulation and the formation of tertiary lymphoid structures in the lung. In both cases immune cells fail to migrate to the lymphatics, leading to highly compromised LN reactions. Mechanistically, ILC2-derived LIF induces the production of the chemokine CCL21 from lymphatic endothelial cells lining the pulmonary lymphatic vessels, thus licensing the homing of CCR7+ immune cells (including dendritic cells) to LNs. Consequently, ILC2-derived LIF dictates the egress of immune cells from the lungs to regulate tissue-localized versus systemic immunity and the balance between allergen and viral responsiveness in the lungs.
Collapse
Affiliation(s)
- Mayuri Gogoi
- MRC Laboratory of Molecular Biology, Cambridge, UK.
| | | | | | | | | | - Michelle Ko
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | - Victor Chen
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Shi-Lu Luan
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | |
Collapse
|
45
|
Chang YC, Tsai HJ, Huang TY, Su NW, Su YW, Chang YF, Chen CGS, Lin J, Chang MC, Chen SJ, Chen HC, Lim KH, Chang KC, Kuo SH. Analysis of mutation profiles in extranodal NK/T-cell lymphoma: clinical and prognostic correlations. Ann Hematol 2024; 103:2917-2930. [PMID: 38671297 DOI: 10.1007/s00277-024-05698-9] [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: 07/18/2023] [Accepted: 03/03/2024] [Indexed: 04/28/2024]
Abstract
The molecular pathogenesis of extranodal NK/T-cell lymphoma (NKTCL) remains obscured despite the next-generation sequencing (NGS) studies explored on ever larger cohorts in the last decade. We addressed the highly variable mutation frequencies reported among previous studies with comprehensive amplicon coverage and enhanced sequencing depth to achieve higher genomic resolution for novel genetic discovery and comparative mutational profiling of the oncogenesis of NKTCL. Targeted exome sequencing was conducted to interrogate 415 cancer-related genes in a cohort of 36 patients with NKTCL, and a total of 548 single nucleotide variants (SNVs) and 600 Copy number variances (CNVs) were identified. Recurrent amplification of the MCL1 (67%) and PIM1 (56%) genes was detected in a dominant majority of patients in our cohort. Functional mapping of genetic aberrations revealed that an enrichment of mutations in the JAK-STAT signaling pathway, including the cytokine receptor LIFR (copy number loss) upstream of JAK3, STAT3 (activating SNVs), and downstream effectors of MYC, PIM1 and MCL1 (copy number gains). RNA in situ hybridization showed the significant consistence of MCL1 RNA level and copy number of MCL1 gene. We further correlated molecular and clinical parameters with overall survival (OS) of these patients. When correlations were analyzed by univariate followed by multivariate modelling, only copy number loss of LIFR gene and stage (III-IV) were independent prognostic factors of reduced OS. Our findings identified that novel loss of LIFR gene significantly correlated with the adverse clinical outcome of NKTCL patients and provided therapeutic opportunities for this disease through manipulating LIFR.
Collapse
Affiliation(s)
- Yu-Cheng Chang
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - To-Yu Huang
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Nai-Wen Su
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Ying-Wen Su
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
- Department of Nursing, Nursing, and Management, MacKay Junior College of Medicine, New Taipei City, Taiwan
| | - Yi-Fang Chang
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Caleb Gon-Shen Chen
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
- Department of Nursing, Nursing, and Management, MacKay Junior College of Medicine, New Taipei City, Taiwan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Johnson Lin
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
| | - Ming-Chih Chang
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | | | | | - Ken-Hong Lim
- Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, New Taipei City, 10449, Taiwan.
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.
- Laboratory of Good Clinical Research Center, Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.
| | - Kung-Chao Chang
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| | - Sung-Hsin Kuo
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
46
|
Xiao K, Wang S, Chen W, Hu Y, Chen Z, Liu P, Zhang J, Chen B, Zhang Z, Li X. Identification of novel immune-related signatures for keloid diagnosis and treatment: insights from integrated bulk RNA-seq and scRNA-seq analysis. Hum Genomics 2024; 18:80. [PMID: 39014455 PMCID: PMC11251391 DOI: 10.1186/s40246-024-00647-z] [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: 06/01/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Keloid is a disease characterized by proliferation of fibrous tissue after the healing of skin tissue, which seriously affects the daily life of patients. However, the clinical treatment of keloids still has limitations, that is, it is not effective in controlling keloids, resulting in a high recurrence rate. Thus, it is urgent to identify new signatures to improve the diagnosis and treatment of keloids. METHOD Bulk RNA seq and scRNA seq data were downloaded from the GEO database. First, we used WGCNA and MEGENA to co-identify keloid/immune-related DEGs. Subsequently, we used three machine learning algorithms (Randomforest, SVM-RFE, and LASSO) to identify hub immune-related genes of keloid (KHIGs) and investigated the heterogeneous expression of KHIGs during fibroblast subpopulation differentiation using scRNA-seq. Finally, we used HE and Masson staining, quantitative reverse transcription-PCR, western blotting, immunohistochemical, and Immunofluorescent assay to investigate the dysregulated expression and the mechanism of retinoic acid in keloids. RESULTS In the present study, we identified PTGFR, RBP5, and LIF as KHIGs and validated their diagnostic performance. Subsequently, we constructed a novel artificial neural network molecular diagnostic model based on the transcriptome pattern of KHIGs, which is expected to break through the current dilemma faced by molecular diagnosis of keloids in the clinic. Meanwhile, the constructed IG score can also effectively predict keloid risk, which provides a new strategy for keloid prevention. Additionally, we observed that KHIGs were also heterogeneously expressed in the constructed differentiation trajectories of fibroblast subtypes, which may affect the differentiation of fibroblast subtypes and thus lead to dysregulation of the immune microenvironment in keloids. Finally, we found that retinoic acid may treat or alleviate keloids by inhibiting RBP5 to differentiate pro-inflammatory fibroblasts (PIF) to mesenchymal fibroblasts (MF), which further reduces collagen secretion. CONCLUSION In summary, the present study provides novel immune signatures (PTGFR, RBP5, and LIF) for keloid diagnosis and treatment, and identifies retinoic acid as potential anti-keloid drugs. More importantly, we provide a new perspective for understanding the interactions between different fibroblast subtypes in keloids and the remodeling of their immune microenvironment.
Collapse
Affiliation(s)
- Kui Xiao
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Sisi Wang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Wenxin Chen
- Department of Gynaecology and Obstetrics, Hengyang Central Hospital, Hunan Normal University, Hengyang, China
| | - Yiping Hu
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Ziang Chen
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Peng Liu
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Jinli Zhang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Bin Chen
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| | - Zhi Zhang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| | - Xiaojian Li
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
47
|
Speckhart SL, Oliver MA, Keane JA, Dias NW, Mercadante VRG, Biase FH, Ealy AD. Interleukin-6 supplementation improves bovine conceptus elongation and transcriptomic indicators of developmental competence†. Biol Reprod 2024; 111:43-53. [PMID: 38519105 PMCID: PMC11247277 DOI: 10.1093/biolre/ioae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/15/2023] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
A high incidence of pregnancy failures occurs in cattle during the second week of pregnancy as blastocysts transition into an elongated conceptus. This work explored whether interleukin-6 supplementation during in vitro embryo production would improve subsequent conceptus development. Bovine embryos were treated with 0 or 100 ng/mL recombinant bovine interleukin-6 beginning on day 5 post-fertilization. At day 7.5 post-fertilization, blastocysts were transferred into estrus synchronized beef cows (n = 5 recipients/treatment, 10 embryos/recipient). Seven days after transfer (day 14.5), cows were euthanized to harvest reproductive tracts and collect conceptuses. Individual conceptus lengths and stages were recorded before processing for RNA sequencing. Increases in conceptus recovery, length, and the proportion of tubular and filamentous conceptuses were detected in conceptuses derived from interleukin-6-treated embryos. The interleukin-6 treatment generated 591 differentially expressed genes in conceptuses (n = 9-10/treatment). Gene ontology enrichment analyses revealed changes in transcriptional regulation, DNA-binding, and antiviral actions. Only a few differentially expressed genes were associated with extraembryonic development, but several differentially expressed genes were associated with embryonic regulation of transcription, mesoderm and ectoderm development, organogenesis, limb formation, and somatogenesis. To conclude, this work provides evidence that interleukin-6 treatment before embryo transfer promotes pre-implantation conceptus development and gene expression in ways that resemble the generation of a robust conceptus containing favorable abilities to survive this critical period of pregnancy.
Collapse
Affiliation(s)
- Savannah L Speckhart
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Mary A Oliver
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jessica A Keane
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Nicholas W Dias
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Vitor R G Mercadante
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Fernando H Biase
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Alan D Ealy
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
48
|
Alvarez AM, Trufen CEM, Buri MV, de Sousa MBN, Arruda-Alves FI, Lichtenstein F, Castro de Oliveira U, Junqueira-de-Azevedo IDLM, Teixeira C, Moreira V. Tumor Necrosis Factor-Alpha Modulates Expression of Genes Involved in Cytokines and Chemokine Pathways in Proliferative Myoblast Cells. Cells 2024; 13:1161. [PMID: 38995013 PMCID: PMC11240656 DOI: 10.3390/cells13131161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Skeletal muscle regeneration after injury is a complex process involving inflammatory signaling and myoblast activation. Pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) are key mediators, but their effects on gene expression in proliferating myoblasts are unclear. We performed the RNA sequencing of TNF-α treated C2C12 myoblasts to elucidate the signaling pathways and gene networks regulated by TNF-α during myoblast proliferation. The TNF-α (10 ng/mL) treatment of C2C12 cells led to 958 differentially expressed genes compared to the controls. Pathway analysis revealed significant regulation of TNF-α signaling, along with the chemokine and IL-17 pathways. Key upregulated genes included cytokines (e.g., IL-6), chemokines (e.g., CCL7), and matrix metalloproteinases (MMPs). TNF-α increased myogenic factor 5 (Myf5) but decreased MyoD protein levels and stimulated the release of MMP-9, MMP-10, and MMP-13. TNF-α also upregulates versican and myostatin mRNA. Overall, our study demonstrates the TNF-α modulation of distinct gene expression patterns and signaling pathways that likely contribute to enhanced myoblast proliferation while suppressing premature differentiation after muscle injury. Elucidating the mechanisms involved in skeletal muscle regeneration can aid in the development of regeneration-enhancing therapeutics.
Collapse
Affiliation(s)
- Angela María Alvarez
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Reproduction Group, Pharmacy Department, School of Pharmaceutical and Food Sciences, University of Antioquia—UdeA, Medellín 050010, Colombia
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| | - Carlos Eduardo Madureira Trufen
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, v.i, 252 50 Vestec, Czech Republic
| | - Marcus Vinicius Buri
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Marcela Bego Nering de Sousa
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| | - Francisco Ivanio Arruda-Alves
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Flavio Lichtenstein
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Ursula Castro de Oliveira
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (U.C.d.O.); (I.d.L.M.J.-d.-A.)
| | | | - Catarina Teixeira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Laboratório de Farmacologia, Butantan Institute, Sao Paulo 05503-900, SP, Brazil
| | - Vanessa Moreira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| |
Collapse
|
49
|
Geethadevi A, Ku Z, Tsaih SW, Parashar D, Kadamberi IP, Xiong W, Deng H, George J, Kumar S, Mittal S, Zhang N, Pradeep S, An Z, Chaluvally-Raghavan P. Blocking Oncostatin M receptor abrogates STAT3 mediated integrin signaling and overcomes chemoresistance in ovarian cancer. NPJ Precis Oncol 2024; 8:127. [PMID: 38839865 PMCID: PMC11153533 DOI: 10.1038/s41698-024-00593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Chemotherapy such as cisplatin is widely used to treat ovarian cancer either before or after surgical debulking. However, cancer relapse due to chemotherapy resistance is a major challenge in the treatment of ovarian cancer. The underlying mechanisms related to chemotherapy resistance remain largely unclear. Therefore, identification of effective therapeutic strategies is urgently needed to overcome therapy resistance. Transcriptome-based analysis, in vitro studies and functional assays identified that cisplatin-resistant ovarian cancer cells express high levels of OSMR compared to cisplatin sensitive cells. Furthermore, OSMR expression associated with a module of integrin family genes and predominantly linked with integrin αV (ITGAV) and integrin β3 (ITGB3) for cisplatin resistance. Using ectopic expression and knockdown approaches, we proved that OSMR directly regulates ITGAV and ITGB3 gene expression through STAT3 activation. Notably, targeting OSMR using anti-OSMR human antibody inhibited the growth and metastasis of ovarian cancer cells and sensitized cisplatin treatment. Taken together, our results underscore the pivotal role of OSMR as a requirement for cisplatin resistance in ovarian cancer. Notably, OSMR fostered the expression of a distinct set of integrin genes, which in turn resulted into a crosstalk between OSMR and integrins for signaling activation that is critical for cisplatin resistance. Therefore, targeting OSMR emerges as a promising and viable strategy to reverse cisplatin-resistance in ovarian cancer.
Collapse
Affiliation(s)
- Anjali Geethadevi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ishaque P Kadamberi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Jasmine George
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sudhir Kumar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sonam Mittal
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA.
| | - Pradeep Chaluvally-Raghavan
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
50
|
Hubel E, Neumann A, Fishman S, Schaffer O, Erez N, Shrkihe BA, Shteingard Y, Gross T, Shibolet O, Varol C, Zvibel I. Sortilin in Biliary Epithelial Cells Promotes Ductular Reaction and Fibrosis during Cholestatic Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:941-957. [PMID: 38493927 DOI: 10.1016/j.ajpath.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
Cholestatic injuries are accompanied by ductular reaction, initiated by proliferation and activation of biliary epithelial cells (BECs), leading to fibrosis. Sortilin (encoded by Sort1) facilitates IL-6 secretion and leukemia inhibitory factor (LIF) signaling. This study investigated the interplay between sortilin and IL-6 and LIF in cholestatic injury-induced ductular reaction, morphogenesis of new ducts, and fibrosis. Cholestatic injury was induced by bile duct ligation (BDL) in wild-type and Sort1-/- mice, with or without augmentation of IL-6 or LIF. Mice with BEC sortilin deficiency (hGFAPcre.Sort1fl/fl) and control mice were subjected to BDL and 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet (DDC) induced cholestatic injury. Sort1-/- mice displayed reduced BEC proliferation and expression of BEC-reactive markers. Administration of LIF or IL-6 restored BEC proliferation in Sort1-/- mice, without affecting BEC-reactive or inflammatory markers. Sort1-/- mice also displayed impaired morphogenesis, which was corrected by LIF treatment. Similarly, hGFAPcre.Sort1fl/fl mice exhibited reduced BEC proliferation, but similar reactive and inflammatory marker expression. Serum IL-6 and LIF were comparable, yet liver pSTAT3 was reduced, indicating that sortilin is essential for co-activation of LIF receptor/gp130 signaling in BECs, but not for IL-6 secretion. hGFAPcre.Sortfl/fl mice displayed impaired morphogenesis and diminished fibrosis after BDL and DDC. In conclusion, sortilin-mediated engagement of LIF signaling in BECs promoted ductular reaction and morphogenesis during cholestatic injury. This study indicates that BEC sortilin is pivotal for the development of fibrosis.
Collapse
Affiliation(s)
- Einav Hubel
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anat Neumann
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sigal Fishman
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ortal Schaffer
- Department of Pediatric Surgery, Assaf Harofe Hospital, Tzrifin, Israel
| | - Noam Erez
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Bander Abu Shrkihe
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Shteingard
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tamar Gross
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Isabel Zvibel
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|