1
|
Lee K, Kim K, Kim JY, Kim JW, Kang YH, Kim YH, Kim SJ. Mechanisms Underlying Medication-Related Osteonecrosis of the Jaw. Oral Dis 2025; 31:1073-1083. [PMID: 39552606 DOI: 10.1111/odi.15198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVE Medication-related osteonecrosis of the jaw (MRONJ) is a rare but debilitating disease characterized by a progressive necrosis of jaw bones in patients who have received anti-resorptive or anti-angiogenic therapies. Unfortunately, we still have no validated preventive or pharmaceutical interventions to help these patients, primarily due to our limited understanding of MRONJ pathogenesis. Here, we offer an extensive review of recent studies relevant to MRONJ pathogenesis. We present a hypothesis regarding the coupling of bone resorption and angiogenesis that relies on osteoblast-derived, matrix-bound vascular endothelial growth factors to explain why ONJ is associated with both anti-resorptive and anti-angiogenic agents. METHODS A narrative review was conducted by searching databases, including PubMed, Scopus, Google Scholar, and Web of Science, to retrieve relevant reports. RESULTS Reduced bone resorption leads to reduced angiogenesis, and vice versa, creating a vicious cycle that ultimately results in ischemic necrosis of the jaw. Additionally, we suggest that reduced angiogenesis, induced by anti-resorptive or anti-angiogenic agents, aggravates bacterial infection-induced bone necrosis, explaining why the jaw bone is particularly susceptible to necrosis. CONCLUSION Our novel hypothesis will facilitate the advancement of future research and the development of more targeted approaches to managing MRONJ.
Collapse
Affiliation(s)
- Kyeongho Lee
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Kihun Kim
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - June Yeon Kim
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Jin-Woo Kim
- Department of Oral and Maxillofacial Surgery, Research Institute for Intractable Osteonecrosis of the Jaw, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Young-Hoon Kang
- Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Yun Hak Kim
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sung-Jin Kim
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
2
|
Gao R, Wu Z, Dang W, Yang T, Chen J, Cheng H, Cui J, Lin L, Shen X, Li F, Yan J, Gao Y, Gao Y, Ma Z. Th1/Th2 Immune Imbalance in the Spleen of Mice Induced by Hypobaric Hypoxia Stimulation and Therapeutic Intervention of Astragaloside IV. Int J Mol Sci 2025; 26:2584. [PMID: 40141225 PMCID: PMC11942621 DOI: 10.3390/ijms26062584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
This study aims to establish a hypobaric hypoxia-induced immune injury model and investigate the intervention and therapeutic effects of Astragaloside IV (AS-IV). This study simulated hypobaric hypoxia stimulation in mice at an altitude of 7000 m on a plateau for 1, 3, 5, and 7 days. HE staining and transcriptomic analysis were performed on mouse spleens. In addition, AS-IV was selected for intervention in prevention and treatment, and validated by flow cytometry, ELISA, and Q-PCR. The results showed that under simulated hypoxic conditions at an altitude of 7000 m for 5 days, the peripheral blood lymphocytes of mice decreased, and the CD45+ cells, CD3+ T cells, and CD3+CD4+ T cells, and CD4+/CD8+ cell ratio in the spleen all decreased. AS-IV can significantly alleviate pathological damage to the spleen, decrease serum levels of IL-2 and IL-6, increase IL-4 and IL-10, and raise CD3+CD4+ T cells and the CD4+/CD8+ cell ratio in peripheral blood and the spleen, while increasing CD4+IFN-γ+cells in spleen, reducing ROS and apoptosis levels in spleen, and increasing the content of relevant mRNA in the Th1/Th2 cell pathway. In summary, simulating hypoxia at an altitude of 7000 m for 5 days can establish a stable hypobaric hypoxic immune injury model, and AS-IV can effectively alleviate hypobaric hypoxic immune injury.
Collapse
Affiliation(s)
- Rong Gao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (R.G.); (W.D.); (J.C.); (H.C.); (F.L.)
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Zhenhui Wu
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330000, China
| | - Wanyun Dang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (R.G.); (W.D.); (J.C.); (H.C.); (F.L.)
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Tingyu Yang
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Junru Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (R.G.); (W.D.); (J.C.); (H.C.); (F.L.)
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Hongbo Cheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (R.G.); (W.D.); (J.C.); (H.C.); (F.L.)
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Jialu Cui
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Lin
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin Shen
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Fangyang Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (R.G.); (W.D.); (J.C.); (H.C.); (F.L.)
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Jiayi Yan
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yehui Gao
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Gao
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| | - Zengchun Ma
- Beijing Institute of Radiation Medicine, Beijing 100859, China; (Z.W.); (T.Y.); (J.C.); (L.L.); (X.S.); (J.Y.); (Y.G.)
| |
Collapse
|
3
|
Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16:1487296. [PMID: 40092992 PMCID: PMC11906415 DOI: 10.3389/fimmu.2025.1487296] [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: 08/27/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The emergence of immunotherapies has revolutionized cancer treatment by leveraging the immune system to target malignancies, offering new hope where traditional therapies often fall short. Within this context, hyperthermia (HT) has re-emerged as a promising adjunctive treatment, capable of enhancing the effectiveness of radiotherapy, chemotherapy, and immunotherapy. HT influences both the innate and adaptive immune systems, enhancing the activity of immune cells such as neutrophils, NK cells, and dendritic cells, while also modulating the tumor microenvironment (TME) to promote immunogenic cell death (ICD) and reduce immunosuppressive conditions. These effects contribute to the transformation of immunologically "cold" tumors into "hot" tumors, making them more susceptible to immune-mediated destruction. Furthermore, HT can amplify the efficacy of immune checkpoint inhibitors (ICIs) by improving immune cell infiltration, inducing damage-associated molecular pattern (DAMP) release, and enhancing antigen presentation. Preclinical and clinical studies support the combination of HT with ICIs, demonstrating improved outcomes in otherwise resistant tumors. However, the full therapeutic potential of the different technologies allowing to apply HT remains to be fully understood, and further research is needed to optimize treatment protocols, explore the differential impacts of local versus whole-body hyperthermia, and identify biomarkers for patient stratification. This review underscores the multifaceted role of HT in immunity and its potential to significantly enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- M Marc Abreu
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, BTT Medical Institute, Aventura, FL, United States
| | - Alberto F Chocron
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- Research Service, Miami Veteran Administration Medical Center, Miami, FL, United States
| | - David M Smadja
- Department of Hematology, AP-HP, Georges Pompidou European Hospital, Paris, France
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
4
|
Senanayake IM, Alam S, Kabir MSH, Petrilla AF, Siraj Z, Theis T, Chekmenev EY, Goodson BM. 15N SABRE-SHEATH and NMR/DFT Characterization of Amino-Metronidazole, a Metabolic Product of the Antibiotic and Prospective Hypoxia Contrast Agent Metronidazole. J Phys Chem B 2025; 129:1662-1669. [PMID: 39843250 PMCID: PMC11969678 DOI: 10.1021/acs.jpcb.4c07877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The antibiotic metronidazole (MNZ) has gained interest as a potential MRI contrast agent for imaging hypoxia. 15N-labeled MNZ can be efficiently hyperpolarized via SABRE-SHEATH (Signal Amplification By Reversible Exchange in SHield Enables Alignment Transfer to Heteronuclei), but the envisioned MRI approach requires that MNZ rapidly undergoes structural changes in hypoxic environments with significant 15N frequency differences manifested in its downstream metabolic products. We have performed NMR studies of the anticipated metabolic product amino-MNZ (despite anticipated stability concerns) accompanied by computational density functional theory (DFT) studies to predict the 15N chemical shifts of different relevant species. Direct hyperpolarization of sparse naturally abundant 15N spins in amino-MNZ via SABRE-SHEATH (enhancement up to ∼9400 fold), along with 1H-decoupled 15N NMR, allowed comparison with both 15N3-MNZ and naturally abundant MNZ. The results show significant 15N shift differences that agree with the DFT predictions. Taken together, the results show that it should be possible to readily distinguish the parent MNZ from product amino-MNZ in envisioned MRI approaches at clinically relevant magnetic fields.
Collapse
Affiliation(s)
- Ishani M. Senanayake
- School of Chemical & Biomolecular Sciences, Southern Illinois University, Carbondale, Illinois, USA
| | - Shahabuddin Alam
- School of Chemical & Biomolecular Sciences, Southern Illinois University, Carbondale, Illinois, USA
| | - Mohammad S. H. Kabir
- Department of Chemistry, Karmanos Cancer Institute (KCI), Integrative Biosciences (Ibio), Wayne State University, Detroit, Michigan, USA
| | - Anthony F. Petrilla
- School of Chemical & Biomolecular Sciences, Southern Illinois University, Carbondale, Illinois, USA
| | - Zahid Siraj
- School of Chemical & Biomolecular Sciences, Southern Illinois University, Carbondale, Illinois, USA
| | - Thomas Theis
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - Eduard Y. Chekmenev
- Department of Chemistry, Karmanos Cancer Institute (KCI), Integrative Biosciences (Ibio), Wayne State University, Detroit, Michigan, USA
| | - Boyd M. Goodson
- School of Chemical & Biomolecular Sciences, Southern Illinois University, Carbondale, Illinois, USA
| |
Collapse
|
5
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [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: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
6
|
Griffin KV, Saunders MN, Lyssiotis CA, Shea LD. Engineering immunity using metabolically active polymeric nanoparticles. Trends Biotechnol 2024:S0167-7799(24)00345-7. [PMID: 39732608 DOI: 10.1016/j.tibtech.2024.11.016] [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: 08/27/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/30/2024]
Abstract
Immune system functions play crucial roles in both health and disease, and these functions are regulated by their metabolic programming. The field of immune engineering has emerged to develop therapeutic strategies, including polymeric nanoparticles (NPs), that can direct immune cell phenotype and function by directing immunometabolic changes. Precise control of bioenergetic processes may offer the opportunity to prevent undesired immune activity and improve disease-specific outcomes. In this review we discuss the role that polymeric NPs can play in shaping immunometabolism and subsequent immune system activity through particle-mediated delivery of metabolically active agents as either structural components or cargo.
Collapse
Affiliation(s)
- Kate V Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Luscombe C, Jones E, Gregorova M, Jones N, Rivino L. Impact of cryopreservation on immune cell metabolism as measured by SCENITH. OXFORD OPEN IMMUNOLOGY 2024; 6:iqae015. [PMID: 39906176 PMCID: PMC11790226 DOI: 10.1093/oxfimm/iqae015] [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: 05/27/2024] [Revised: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 02/06/2025] Open
Abstract
The dynamic functioning of immune cells is regulated by cellular metabolic processes, and there is growing interest in the study of immunometabolic correlates of dysfunctional immune responses. SCENITH is a novel flow cytometry-based technique that allows for ex vivo metabolic profiling of immune cells within heterogeneous samples. Cryopreservation of clinical samples is frequently undertaken to facilitate high throughput processing and longitudinal analyses of immune responses, but is thought to lead to cellular metabolic dysfunction. We aimed to investigate the impact of cryopreservation on immune cell metabolism, harnessing SCENITH's unique ability to describe the divergent bioenergetic characteristics of distinct immune cell subsets. We demonstrate that upon activation, T cells are unable to sufficiently/readily undergo metabolic reprogramming. Additionally, we find that cryopreservation introduces a time-dependent metabolic artefact that favours glycolysis and impairs oxidative phosphorylation, suggesting that cryopreservation results in mitochondrial dysfunction. Despite this artefact, SCENITH was still able to reveal the distinct bioenergetic profiles of contrasting immune cells populations following cryopreservation. Whilst SCENITH can provide valuable information about immune cell metabolism even in cryopreserved samples, our findings have important implications for the design of future studies. Investigators should carefully consider how to process and store clinical samples to ensure that cryopreservation does not confound analyses, particularly where longitudinal sampling is required.
Collapse
Affiliation(s)
- Curtis Luscombe
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Eben Jones
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Michaela Gregorova
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, United Kingdom
| | - Laura Rivino
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| |
Collapse
|
8
|
Qi J, Gao Y, Chen R, Meng X, Wang L, Zhou M, Yin P, Kan H. Criteria air pollutants and HIV-related mortality: Insights from a nationwide case-crossover investigation. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136249. [PMID: 39490161 DOI: 10.1016/j.jhazmat.2024.136249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
The heightened vulnerability of individuals with HIV to environmental stressors is well-recognized, yet the role of air pollution in exacerbating HIV-related mortality remains underexplored. In this nationwide, individual-level case-crossover study conducted from 2013 to 2019, we investigated the association between short-term exposure to criteria air pollutants and HIV-related mortality. Our analysis of 38,510 HIV-related deaths revealed significant associations between exposure to PM2.5, PM10, NO2, and CO and increased mortality risk. The effects of PM2.5 and PM10 persisted for two days, whereas NO2 and CO had immediate, same-day impacts. Vulnerability was heightened in individuals under 65 years, males, those with lower educational attainment, and unmarried individuals. Among causes of death, HIV-related malignant neoplasms exhibited the highest sensitivity to particulate matter. Our findings provide novel insights into the relationship between short-term air pollution exposure and HIV-related mortality, emphasizing the increased susceptibility of this immunocompromised population. The results underscore the need for targeted public health interventions to reduce pollution exposure, particularly for the most at-risk demographic groups. This study contributes to a deeper understanding of environmental health risks faced by individuals living with HIV and informs evidence-based policy recommendations.
Collapse
Affiliation(s)
- Jinlei Qi
- National Center for Chronic Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ya Gao
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Renjie Chen
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Xia Meng
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Lijun Wang
- National Center for Chronic Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Maigeng Zhou
- National Center for Chronic Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peng Yin
- National Center for Chronic Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Haidong Kan
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China; Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China.
| |
Collapse
|
9
|
Di Mattia M, Sallese M, Neri M, Lopetuso LR. Hypoxic Functional Regulation Pathways in the GI Tract: Focus on the HIF-1α and Microbiota's Crosstalk. Inflamm Bowel Dis 2024; 30:1406-1418. [PMID: 38484200 DOI: 10.1093/ibd/izae046] [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: 07/28/2023] [Indexed: 08/02/2024]
Abstract
Hypoxia is an essential gastrointestinal (GI) tract phenomenon that influences both physiologic and pathologic states. Hypoxia-inducible factors (HIFs), the primary drivers of cell adaptation to low-oxygen environments, have been identified as critical regulators of gut homeostasis: directly, through the induction of different proteins linked to intestinal barrier stabilization (ie, adherent proteins, tight junctions, mucins, integrins, intestinal trefoil factor, and adenosine); and indirectly, through the regulation of several immune cell types and the modulation of autophagy and inflammatory processes. Furthermore, hypoxia and HIF-related sensing pathways influence the delicate relationship existing between bacteria and mammalian host cells. In turn, gut commensals establish and maintain the physiologic hypoxia of the GI tract and HIF-α expression. Based on this premise, the goals of this review are to (1) highlight hypoxic molecular pathways in the GI tract, both in physiologic and pathophysiologic settings, such as inflammatory bowel disease; and (2) discuss a potential strategy for ameliorating gut-related disorders, by targeting HIF signaling, which can alleviate inflammatory processes, restore autophagy correct mechanisms, and benefit the host-microbiota equilibrium.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Matteo Neri
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
10
|
Wirz K, Schulz C, Söbbeler F, Armando F, Beythien G, Gerhauser I, de Buhr N, Pilchová V, Meyer zu Natrup C, Baumgärtner W, Kästner S, von Köckritz-Blickwede M. A New Methodology for the Oxygen Measurement in Lung Tissue of an Aged Ferret Model Proves Hypoxia during COVID-19. Am J Respir Cell Mol Biol 2024; 71:146-153. [PMID: 39087829 PMCID: PMC11299086 DOI: 10.1165/rcmb.2024-0005ma] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/30/2024] [Indexed: 08/02/2024] Open
Abstract
Oxygen as a key element has a high impact on cellular processes. Infection with a pathogen such as SARS-CoV-2 and after inflammation may lead to hypoxic conditions in tissue that impact cellular responses. To develop optimized translational in vitro models for a better understanding of physiologic and pathophysiologic oxygen conditions, it is a prerequisite to determine oxygen concentrations generated in vivo. Our study objective was the establishment of an invasive method for oxygen measurements using a luminescence-based microsensor to determine the dissolved oxygen in the lung tissue of ferrets as animal models for SARS-CoV-2 research. By way of analogy to humans, aged ferrets are more likely to show clinical signs after SARS-CoV-2 infection than are young animals. To investigate oxygen concentrations during a respiratory viral infection, we intratracheally infected nine aged (3-yr-old) ferrets with SARS-CoV-2. The aged SARS-CoV-2-infected ferrets showed mild to moderate clinical signs associated with prolonged viral RNA shedding until 14 days postinfection. SARS-CoV-2-infected ferrets showed histopathologic lung lesion scores that significantly negatively correlated with oxygen concentrations in lung tissue. At 4 days postinfection, oxygen concentrations in lung tissue were significantly lower (mean percentage O2, 3.89 ≙ ≈ 27.78 mm Hg) than in the negative control group (mean percentage O2, 8.65 ≙ ≈ 61.4 mm Hg). In summary, we succeeded in determining the pathophysiologic oxygen conditions in the lung tissue of aged SARS-CoV-2-infected ferrets.
Collapse
Affiliation(s)
- Katrin Wirz
- Research Center for Emerging Infections and Zoonoses
- Institute of Biochemistry
| | | | | | | | | | | | - Nicole de Buhr
- Research Center for Emerging Infections and Zoonoses
- Institute of Biochemistry
| | - Veronika Pilchová
- Research Center for Emerging Infections and Zoonoses
- Institute of Biochemistry
| | - Christian Meyer zu Natrup
- Research Center for Emerging Infections and Zoonoses
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | | | | |
Collapse
|
11
|
Qin Y, Zhang X, Chen Y, Zhang W, Du S, Ren C. Prognostic Analysis of a Hypoxia-Associated lncRNA Signature in Glioblastoma and its Pan-Cancer Landscape. J Neurol Surg A Cent Eur Neurosurg 2024; 85:378-388. [PMID: 37023792 DOI: 10.1055/a-2070-3715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
BACKGROUND Hypoxia is an important clinical feature of glioblastoma (GBM), which regulates a variety of tumor processes and is inseparable from radiotherapy. Accumulating evidence suggests that long noncoding RNAs (lncRNAs) are strongly associated with survival outcomes in GBM patients and modulate hypoxia-induced tumor processes. Therefore, the aim of this study was to establish a hypoxia-associated lncRNAs (HALs) prognostic model to predict survival outcomes in GBM patients. METHODS LncRNAs in GBM samples were extracted from The Cancer Genome Atlas database. Hypoxia-related genes were downloaded from the Molecular Signature Database. Co-expression analysis of differentially expressed lncRNAs and hypoxia-related genes in GBM samples was performed to determine HALs. Six optimal lncRNAs were selected for building HALs models by univariate Cox regression analysis. RESULTS The prediction model has a good predictive effect on the prognosis of GBM patients. Meanwhile, LINC00957 among the six lncRNAs was selected and subjected to pan-cancer landscape analysis. CONCLUSION Taken together, our findings suggest that the HALs assessment model can be used to predict the prognosis of GBM patients. In addition, LINC00957 included in the model may be a useful target to study the mechanism of cancer development and design individualized treatment strategies.
Collapse
Affiliation(s)
- Yue Qin
- Department of Radiation Oncology, Southern Medical University, Guangzhou, China
| | - Xiaonan Zhang
- Department of Radiation Oncology, Southern Medical University, Guangzhou, China
| | - Yulei Chen
- Department of Radiation Oncology, Southern Medical University, Guangzhou, China
| | - Wan Zhang
- Department of Radiation Oncology, Southern Medical University, Guangzhou, China
| | - Shasha Du
- Department of Radiation Oncology, Guangdong Provincial People's Hospital, Guangzhou, Guangdong, China
| | - Chen Ren
- Department of Radiation Oncology, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Xiu W, Dong H, Chen X, Wan L, Lu L, Yang K, Yuwen L, Li Q, Ding M, Zhang Y, Mou Y, Wang L. Metabolic Modulation-Mediated Antibiotic and Immune Activation for Treatment of Chronic Lung Infections. ACS NANO 2024; 18:15204-15217. [PMID: 38803167 DOI: 10.1021/acsnano.4c03527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The Pseudomonas aeruginosa biofilm in recalcitrant chronic lung infections not only develops high antimicrobial tolerance but also induces an aberrant host inflammatory response. The metabolic condition plays a vital role in both the antimicrobial susceptibility of bacteria and the inflammatory response of immune cells, thereby offering a potential therapeutic target. Herein, we described a metabolic modulation strategy by using ultrasound-responsive liposomal nanoparticles containing a sonosensitizer and a hypoxia-activated prodrug against biofilm-associated chronic lung infections. Under ultrasound stimulation, the sonosensitizer generates antibacterial reactive oxygen species by oxygen consumption. Subsequently, the oxygen consumption-mediated hypoxia not only induces the anaerobic metabolism of bacteria for antibiotic activation but also triggers the glycolysis pathway of immune cells for inflammatory activation. Such metabolic modulation strategy demonstrated efficient therapeutic efficacy for P. aeruginosa biofilm-induced chronic lung infections in mice models and provides a promising way for combating biofilm-associated chronic infections.
Collapse
Affiliation(s)
- Weijun Xiu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Heng Dong
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaolong Chen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Ling Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Liang Lu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Kaili Yang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Lihui Yuwen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yu Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| |
Collapse
|
13
|
Montúfar-Romero M, Valenzuela-Muñoz V, Valenzuela-Miranda D, Gallardo-Escárate C. Hypoxia in the Blue Mussel Mytilus chilensis Induces a Transcriptome Shift Associated with Endoplasmic Reticulum Stress, Metabolism, and Immune Response. Genes (Basel) 2024; 15:658. [PMID: 38927594 PMCID: PMC11203016 DOI: 10.3390/genes15060658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The increase in hypoxia events, a result of climate change in coastal and fjord ecosystems, impacts the health and survival of mussels. These organisms deploy physiological and molecular responses as an adaptive mechanism to maintain cellular homeostasis under environmental stress. However, the specific effects of hypoxia on mussels of socioeconomic interest, such as Mytilus chilensis, are unknown. Using RNA-seq, we investigated the transcriptomic profiles of the gills, digestive gland, and adductor muscle of M. chilensis under hypoxia (10 days at 2 mg L-1) and reoxygenation (10 days at 6 mg L-1). There were 15,056 differentially expressed transcripts identified in gills, 11,864 in the digestive gland, and 9862 in the adductor muscle. The response varied among tissues, showing chromosomal changes in Chr1, Chr9, and Chr10 during hypoxia. Hypoxia regulated signaling genes in the Toll-like, mTOR, citrate cycle, and apoptosis pathways in gills, indicating metabolic and immunological alterations. These changes suggest that hypoxia induced a metabolic shift in mussels, reducing reliance on aerobic respiration and increasing reliance on anaerobic metabolism. Furthermore, hypoxia appeared to suppress the immune response, potentially increasing disease susceptibility, with negative implications for the mussel culture industry and natural bed populations. This study provides pivotal insights into metabolic and immunological adaptations to hypoxia in M. chilensis, offering candidate genes for adaptive traits.
Collapse
Affiliation(s)
- Milton Montúfar-Romero
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (D.V.-M.)
- Biotecnology Center, Universidad de Concepción, Concepción 4030000, Chile
- Instituto Público de Investigación de Acuicultura y Pesca (IPIAP), Guayaquil 090314, Ecuador
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (D.V.-M.)
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Naturaleza, Universidad San Sebastián, Concepción 4030000, Chile
| | - Diego Valenzuela-Miranda
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (D.V.-M.)
- Biotecnology Center, Universidad de Concepción, Concepción 4030000, Chile
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (D.V.-M.)
- Biotecnology Center, Universidad de Concepción, Concepción 4030000, Chile
| |
Collapse
|
14
|
Allsopp GL, Addinsall AB, Stephenson G, Basheer F, Gatta PAD, Hoffmann SM, Russell AP, Wright CR. The chronic leukocyte and inflammatory cytokine responses of older adults to resistance training in normobaric hypoxia; a randomized controlled trial. BMC Sports Sci Med Rehabil 2024; 16:102. [PMID: 38698481 PMCID: PMC11067184 DOI: 10.1186/s13102-024-00890-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
TRIAL DESIGN Older adults experience chronic dysregulation of leukocytes and inflammatory cytokines, both at rest and in response to resistance training. Systemic hypoxia modulates leukocytes and cytokines, therefore this study characterized the effects of normobaric hypoxia on the leukocyte and cytokine responses of older adults to resistance training. METHODS 20 adults aged 60-70 years performed eight weeks of moderate-intensity resistance training in either normoxia or normobaric hypoxia (14.4% O2), consisting of two lower body and two upper body exercises. Venous blood was drawn before and after the training intervention and flow cytometry was used to quantify resting neutrophils, lymphocytes, monocytes, eosinophils and basophils, in addition to the subsets of lymphocytes (T, B and natural killer (NK) cells). Inflammatory cytokines were also quantified; interleukin 1 beta (IL-1β), IL-4, IL-6, IL-8, IL-10 and tumor necrosis factor alpha (TNF-α). Acute changes in leukocytes and cytokines were also measured in the 24 h following the last training session. RESULTS After the intervention there was a greater concentration of resting white blood cells (p = 0.03; 20.3% higher) T cells (p = 0.008; 25.4% higher), B cells (p = 0.004; 32.6% higher), NK cells (p = 0.012; 43.9% higher) and eosinophils (p = 0.025; 30.8% higher) in hypoxia compared to normoxia, though the cytokines were unchanged. No acute effect of hypoxia was detected in the 24 h following the last training session for any leukocyte population or inflammatory cytokine (p < 0.05). CONCLUSIONS Hypoxic training caused higher concentrations of resting lymphocytes and eosinophils, when compared to normoxic training. Hypoxia may have an additional beneficial effect on the immunological status of older adults. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry (ANZCTR). TRIAL NUMBER ACTRN12623001046695. Registered 27/9/2023. Retrospectively registered. All protocols adhere to the COSORT guidelines.
Collapse
Affiliation(s)
- Giselle Larissa Allsopp
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia.
| | - Alex Bernard Addinsall
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
- Department of Physiology and Pharmacology, Karolinska Insitutet, Stockholm, 171 77, Sweden
| | - Garth Stephenson
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Faiza Basheer
- School of Medicine, Deakin University, Geelong, Victoria, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Victoria, Australia
| | - Paul Adrian Della Gatta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Samantha May Hoffmann
- Centre for Sport Research (CSR), School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Aaron Paul Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Craig Robert Wright
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| |
Collapse
|
15
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
16
|
Wolfschmitt EM, Vogt JA, Hogg M, Wachter U, Stadler N, Kapapa T, Datzmann T, Messerer DAC, Hoffmann A, Gröger M, Münz F, Mathieu R, Mayer S, Merz T, Asfar P, Calzia E, Radermacher P, Zink F. 13C-Metabolic flux analysis detected a hyperoxemia-induced reduction of tricarboxylic acid cycle metabolism in granulocytes during two models of porcine acute subdural hematoma and hemorrhagic shock. Front Immunol 2024; 14:1319986. [PMID: 38332911 PMCID: PMC10850868 DOI: 10.3389/fimmu.2023.1319986] [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: 10/11/2023] [Accepted: 12/15/2023] [Indexed: 02/10/2024] Open
Abstract
Introduction Supplementation with increased inspired oxygen fractions has been suggested to alleviate the harmful effects of tissue hypoxia during hemorrhagic shock (HS) and traumatic brain injury. However, the utility of therapeutic hyperoxia in critical care is disputed to this day as controversial evidence is available regarding its efficacy. Furthermore, in contrast to its hypoxic counterpart, the effect of hyperoxia on the metabolism of circulating immune cells remains ambiguous. Both stimulating and detrimental effects are possible; the former by providing necessary oxygen supply, the latter by generation of excessive amounts of reactive oxygen species (ROS). To uncover the potential impact of increased oxygen fractions on circulating immune cells during intensive care, we have performed a 13C-metabolic flux analysis (MFA) on PBMCs and granulocytes isolated from two long-term, resuscitated models of combined acute subdural hematoma (ASDH) and HS in pigs with and without cardiovascular comorbidity. Methods Swine underwent resuscitation after 2 h of ASDH and HS up to a maximum of 48 h after HS. Animals received normoxemia (PaO2 = 80 - 120 mmHg) or targeted hyperoxemia (PaO2 = 200 - 250 mmHg for 24 h after treatment initiation, thereafter PaO2 as in the control group). Blood was drawn at time points T1 = after instrumentation, T2 = 24 h post ASDH and HS, and T3 = 48 h post ASDH and HS. PBMCs and granulocytes were isolated from whole blood to perform electron spin resonance spectroscopy, high resolution respirometry and 13C-MFA. For the latter, we utilized a parallel tracer approach with 1,2-13C2 glucose, U-13C glucose, and U-13C glutamine, which covered essential pathways of glucose and glutamine metabolism and supplied redundant data for robust Bayesian estimation. Gas chromatography-mass spectrometry further provided multiple fragments of metabolites which yielded additional labeling information. We obtained precise estimations of the fluxes, their joint credibility intervals, and their relations, and characterized common metabolic patterns with principal component analysis (PCA). Results 13C-MFA indicated a hyperoxia-mediated reduction in tricarboxylic acid (TCA) cycle activity in circulating granulocytes which encompassed fluxes of glutamine uptake, TCA cycle, and oxaloacetate/aspartate supply for biosynthetic processes. We further detected elevated superoxide levels in the swine strain characterized by a hypercholesterolemic phenotype. PCA revealed cell type-specific behavioral patterns of metabolic adaptation in response to ASDH and HS that acted irrespective of swine strains or treatment group. Conclusion In a model of resuscitated porcine ASDH and HS, we saw that ventilation with increased inspiratory O2 concentrations (PaO2 = 200 - 250 mmHg for 24 h after treatment initiation) did not impact mitochondrial respiration of PBMCs or granulocytes. However, Bayesian 13C-MFA results indicated a reduction in TCA cycle activity in granulocytes compared to cells exposed to normoxemia in the same time period. This change in metabolism did not seem to affect granulocytes' ability to perform phagocytosis or produce superoxide radicals.
Collapse
Affiliation(s)
- Eva-Maria Wolfschmitt
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Josef Albert Vogt
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Melanie Hogg
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Ulrich Wachter
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Nicole Stadler
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Thomas Kapapa
- Clinic for Neurosurgery, University Hospital Ulm, Ulm, Germany
| | - Thomas Datzmann
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Andrea Hoffmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Michael Gröger
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Franziska Münz
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
| | - René Mathieu
- Clinic for Neurosurgery, Bundeswehrkrankenhaus, Ulm, Germany
| | - Simon Mayer
- Clinic for Neurosurgery, Bundeswehrkrankenhaus, Ulm, Germany
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
| | - Pierre Asfar
- Département de Médecine Intensive – Réanimation et Médecine Hyperbare, Centre Hospitalier Universitaire, Angers, France
| | - Enrico Calzia
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Fabian Zink
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
17
|
Kuloğlu N, Karabulut D, Kaymak E, Akin AT, Ceylan T, Yıldırım AB, Yakan B. Effect of vitamin B12 on methotrexate-induced cardiotoxicity in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:733-739. [PMID: 38645491 PMCID: PMC11024404 DOI: 10.22038/ijbms.2024.74161.16120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/15/2024] [Indexed: 04/23/2024]
Abstract
Objectives Methotrexate (MTX) is a drug with anti-inflammatory and immunosuppressive effects and is also a folic acid antagonist. Our aim in this study is to determine the molecular mechanisms of cardiotoxicity caused by MTX, a chemotherapeutic drug, and to evaluate the protective effects of vitamin B12 on this toxicity. Materials and Methods A total of 32 rats were used in our study and 4 groups were formed. Control group, Vit B12 group (3 μg/kg B12 for 15 days, IP), MTX group (20 mg/kg MTX single dose on day 8 of the experiment, IP), MTX +Vit B12 group (3 μg/kg, IP ), Vit B12 throughout the 15 days, and a single dose of 20 mg/kg MTX (IP) on day 8 of the experiment. Immunohistochemically, expressions of hypoxia-inducible factor 1α (HIF1-α), vascular endothelial growth factor receptor-2 (VEGFR-2), erythropoietin (EPO), and interleukin-6 (IL-6) were evaluated in the heart tissue. Total catalase (CAT), superoxide dismutase (SOD), and malondialdehyde (MDA) levels were measured in the heart tissue. At the same time, ANP and NT-proBNP levels were measured in the blood serum. Results In the study, the expression of HIF1-α and VEGFR-2 increased significantly in the MTX group, while IL-6 and EPO significantly decreased. At the same time, CAT and SOD levels were significantly decreased and MDA levels increased significantly in the MTX group. While vitamin B12 significantly corrected all these values, it also greatly reduced the increases in ANP and NT-proBNP levels caused by MTX. Conclusion It is important to use Vit B12 before and after MTX administration to replace the folate that MTX has reduced.
Collapse
Affiliation(s)
- Nurhan Kuloğlu
- Healthcare Services Department, Niğde Ömer Halisdemir University, Nigde, Turkey
| | - Derya Karabulut
- Histology-Embryology Department, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Emin Kaymak
- Histology-Embryology Department, Faculty of Medicine, Bozok University, Yozgat, Turkey
| | - Ali Tuğrul Akin
- Department of Medical Biology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Tayfun Ceylan
- Histology-Embryology Department, Faculty of Dentistry, Cappadocia University, Nevşehir, Turkey
| | - Ayşegül Burçin Yıldırım
- Gaziantep Islamic Science and Technology University, Department of Histology-Embryology, Gaziantep, Turkey
| | - Birkan Yakan
- Histology-Embryology Department, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
18
|
Kapnick SM, Martin CA, Jewell CM. Engineering metabolism to modulate immunity. Adv Drug Deliv Rev 2024; 204:115122. [PMID: 37935318 PMCID: PMC10843796 DOI: 10.1016/j.addr.2023.115122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Metabolic programming and reprogramming have emerged as pivotal mechanisms for altering immune cell function. Thus, immunometabolism has become an attractive target area for treatment of immune-mediated disorders. Nonetheless, many hurdles to delivering metabolic cues persist. In this review, we consider how biomaterials are poised to transform manipulation of immune cell metabolism through integrated control of metabolic configurations to affect outcomes in autoimmunity, regeneration, transplant, and cancer. We emphasize the features of nanoparticles and other biomaterials that permit delivery of metabolic cues to the intracellular compartment of immune cells, or strategies for altering signals in the extracellular space. We then provide perspectives on the potential for reciprocal regulation of immunometabolism by the physical properties of materials themselves. Lastly, opportunities for clinical translation are highlighted. This discussion contributes to our understanding of immunometabolism, biomaterials-based strategies for altering metabolic configurations in immune cells, and emerging concepts in this evolving field.
Collapse
Affiliation(s)
- Senta M Kapnick
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA
| | - Corinne A Martin
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S Greene Street, Suite N9E17, Baltimore, MD, USA.
| |
Collapse
|
19
|
Wang CA, Hsieh WC, Hsu TJ, Tsai FJ, Hsu CY. Obstructive sleep apnea increases the risk of herpes zoster and postherpetic neuralgia. Postgrad Med 2024; 136:22-29. [PMID: 38011906 DOI: 10.1080/00325481.2023.2288562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE Diseases associated with chronic pain are typically a major source of stress for patients; and have been linked to herpes zoster (HZ) development. Here, we investigated whether obstructive sleep apnea (OSA) is a potential stressor that increases the risk of HZ and postherpetic neuralgia (PHN) in affected individuals. METHODS The data used in this study were obtained from the National Health Insurance Research Database. The study cohort included patients aged between 20 and 100 years who had OSA during the period from 2000 to 2017 (with tracking completed until 2018). The case group and the control group were matched at a 1:1 ratio on the basis of age, sex, comorbidities, and index year, with patients who had outcomes before the index date being excluded. The outcomes considered in this study were HZ and PHN. The risk of HZ and PHN with and without OSA was calculated, and age, sex, comorbidities, and index year were adjusted for. RESULTS There were 25,211 patients in each group. Patients with OSA had a significantly higher risk of HZ (adjusted hazard ratio [aHR] = 1.22) than those without did. The patients with OSA had also a significantly higher risk of PHN (aHR = 1.36) than those without did. In term of comorbidities, the patients with OSA without (aHR = 1.28) and with (aHR = 1.17) comorbidities had a significantly higher risk of HZ compared with those without OSA. In addition, the patients with OSA but no other comorbidities (aHR = 1.68) had a significantly higher risk of PHN than those without did. CONCLUSION OSA increases the risk of not only HZ but also PHN. Therefore, patients with OSA should be aware of the potential effect of the disease on their stress levels, as well as the increased risk of developing HZ and PHN.
Collapse
Affiliation(s)
- Chih-An Wang
- Division of Respiratory Therapy, Ditmanson Medical Foundation, Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Wen-Che Hsieh
- Department of Chinese Medicine, Ditmanson Medical Foundation, Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Tzu-Ju Hsu
- Management Office for Health Data, Clinical Trial Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Chao-Yu Hsu
- Department of Medical Education, Ditmanson Medical Foundation, Chia-Yi Christian Hospital, Chia-Yi, Taiwan
- Department of Artificial Intelligence and Healthcare Management, Central Taiwan University of Science and Technology, Taichung, Taiwan
- Center for General Education, National Taichung University of Science and Technology, Taichung, Taiwan
- Department of General Education, National Chin-Yi University of Technology, Taichung, Taiwan
| |
Collapse
|
20
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
21
|
Bhatt K, Nukovic A, Colombani T, Bencherif SA. Biomaterial-assisted local oxygenation safeguards the prostimulatory phenotype and functions of human dendritic cells in hypoxia. Front Immunol 2023; 14:1278397. [PMID: 38169677 PMCID: PMC10758617 DOI: 10.3389/fimmu.2023.1278397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Dendritic cells (DCs), professional antigen-presenting cells, function as sentinels of the immune system. DCs initiate and fine-tune adaptive immune responses by presenting antigenic peptides to B and T lymphocytes to mount an effective immune response against cancer and pathogens. However, hypoxia, a condition characterized by low oxygen (O2) tension in different tissues, significantly impacts DC functions, including antigen uptake, activation and maturation, migration, as well as T-cell priming and proliferation. In this study, we employed O2-releasing biomaterials (O2-cryogels) to study the effect of localized O2 supply on human DC phenotype and functions. Our results indicate that O2-cryogels effectively mitigate DC exposure to hypoxia under hypoxic conditions. Additionally, O2-cryogels counteract hypoxia-induced inhibition of antigen uptake and migratory activity in DCs through O2 release and hyaluronic acid (HA) mediated mechanisms. Furthermore, O2-cryogels preserve and restore DC maturation and co-stimulation markers, including HLA-DR, CD86, and CD40, along with the secretion of proinflammatory cytokines in hypoxic conditions. Finally, our findings demonstrate that the supplemental O2 released from the cryogels preserves DC-mediated T-cell priming, ultimately leading to the activation and proliferation of allogeneic CD3+ T cells. This work emphasizes the potential of local oxygenation as a powerful immunomodulatory agent to improve DC activation and functions in hypoxia, offering new approaches for cancer and infectious disease treatments.
Collapse
Affiliation(s)
- Khushbu Bhatt
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Alexandra Nukovic
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| |
Collapse
|
22
|
Shi X, Gao F, Zhao X, Pei C, Zhu L, Zhang J, Li C, Li L, Kong X. Role of HIF in fish inflammation. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109222. [PMID: 37956798 DOI: 10.1016/j.fsi.2023.109222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
The hypoxia-inducing factor (HIF) is a central transcription factor in cellular oxygen sensing and regulation. It is common that the inflammation always appears in many diseases, like infectious diseases in fishes, and the inflammation is often accompanied by hypoxia, as a hallmark of inflammation. Besides coordinating cellular responses to low oxygen, HIF-mediated hypoxia signaling pathway is also crucial for immune responses such as the regulations of innate immune cell phenotype and function, as well as metabolic reprogramming under the inflammation. However, the understanding of the molecular mechanisms by which HIFs regulate the inflammatory response in fish is still very limited. Here, we review the characteristics of HIF as well as its roles in innate immune cells and the infections caused by bacteria and viruses. The regulatory effects of HIF on the metabolic reprogramming of innate immune cells are also discussed and the future research directions are outlooked. This paper will serve as a reference for elucidating the molecular mechanism of HIF regulating inflammation and identifying treatment strategies to target HIF for fish disease.
Collapse
Affiliation(s)
- Xiaowei Shi
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China; Sanquan Medical College, Henan Province, PR China
| | - Feng Gao
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Xianliang Zhao
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Chao Pei
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Lei Zhu
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Jie Zhang
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Chen Li
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Li Li
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China
| | - Xianghui Kong
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Henan Province, PR China.
| |
Collapse
|
23
|
Kosyreva AM, Dzhalilova DS, Tsvetkov IS, Makarova MA, Makarova OV. Ex Vivo Production of IL-1β and IL-10 by Activated Blood Cells of Wistar Rats with Different Resistance to Hypoxia after Systemic Inflammatory Response Syndrome. Bull Exp Biol Med 2023; 176:290-296. [PMID: 38194074 DOI: 10.1007/s10517-024-06010-5] [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: 04/06/2023] [Indexed: 01/10/2024]
Abstract
We studied spontaneous and ex vivo activated cytokine production by blood cells of male Wistar rats with different resistance to hypoxia against the background of an LPS-induced systemic inflammatory response. In rats with low (LR) and high resistance (HR) to hypoxia, the number of leukocytes, granulocytes, and peripheral blood lymphocytes was determined, the levels of spontaneous and stimulated production of IL-1β and IL-10 and their ratio were assessed ex vivo. Against the background of a systemic inflammatory response, only HR animals showed a decrease in spontaneous and stimulated production of IL-1β and spontaneous production of IL-10. The IL-1β/IL-10 ratio decreased only in LR rats during the development of a systemic inflammatory response, while in HR animals, no changes in this indicator were observed. The obtained data suggest a high proinflammatory potential of blood cells in LR rats, which apparently determines the development of a more severe course of the systemic inflammatory response.
Collapse
Affiliation(s)
- A M Kosyreva
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia.
| | - D Sh Dzhalilova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - I S Tsvetkov
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - M A Makarova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - O V Makarova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| |
Collapse
|
24
|
Alzamami A. Implications of single-cell immune landscape of tumor microenvironment for the colorectal cancer diagnostics and therapy. Med Oncol 2023; 40:352. [PMID: 37950801 DOI: 10.1007/s12032-023-02226-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023]
Abstract
Colorectal cancer (CRC) originates from the polyps lining the colon and is among the most common types of cancer. With the increasing popularity of single-cell sequencing technologies, researchers have been able to better understand the immune landscape of colorectal cancer, by analyzing their expression and interactions in detail with the tumor microenvironment (TME) at single-cell level. Since the tumor-immune cell interactions play a critical part in the advancement as well as treatment response in colorectal cancer, the release of inhibitory factors such as T cells are important for recognizing and destroying cancer cells. Such information is vital to identify immunotherapeutic targets for cure and monitoring response to treatments. Therefore, a comprehensive single-cell studies-based overview of key immunogenic agents regulating the TME of CRC is provided in this review. Tumor-associated macrophages can promote tumor growth and resistance to treatment by releasing factors that inhibit the function of other immune cells. Additionally, colorectal cancer cells can express programmed cell death protein 1 and its ligand, which can also inhibit T-cell function. Researchers have found that certain types of immune cells, prominently T cells, natural killer, and dendritic cells, can have a positive impact on the prognosis of colorectal cancer patients. Treatments like immune checkpoint inhibitors and CAR-T therapies that help to release the inhibitory signals from the cancer cells allow the immune cells to function more effectively.
Collapse
Affiliation(s)
- Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Sciences, Shaqra University, 11961, Al-Quwayiyah, Saudi Arabia.
| |
Collapse
|
25
|
Rani B, Ignatz-Hoover JJ, Rana PS, Driscoll JJ. Current and Emerging Strategies to Treat Urothelial Carcinoma. Cancers (Basel) 2023; 15:4886. [PMID: 37835580 PMCID: PMC10571746 DOI: 10.3390/cancers15194886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Urothelial cell carcinoma (UCC, bladder cancer, BC) remains a difficult-to-treat malignancy with a rising incidence worldwide. In the U.S., UCC is the sixth most incident neoplasm and ~90% of diagnoses are made in those >55 years of age; it is ~four times more commonly observed in men than women. The most important risk factor for developing BC is tobacco smoking, which accounts for ~50% of cases, followed by occupational exposure to aromatic amines and ionizing radiation. The standard of care for advanced UCC includes platinum-based chemotherapy and programmed cell death (PD-1) or programmed cell death ligand 1 (PD-L1) inhibitors, administered as frontline, second-line, or maintenance therapy. UCC remains generally incurable and is associated with intrinsic and acquired drug and immune resistance. UCC is lethal in the metastatic state and characterized by genomic instability, high PD-L1 expression, DNA damage-response mutations, and a high tumor mutational burden. Although immune checkpoint inhibitors (ICIs) achieve long-term durable responses in other cancers, their ability to achieve similar results with metastatic UCC (mUCC) is not as well-defined. Here, we discuss therapies to improve UCC management and how comprehensive tumor profiling can identify actionable biomarkers and eventually fulfill the promise of precision medicine for UCC patients.
Collapse
Affiliation(s)
- Berkha Rani
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (B.R.); (J.J.I.-H.); (P.S.R.)
| | - James J. Ignatz-Hoover
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (B.R.); (J.J.I.-H.); (P.S.R.)
- Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Priyanka S. Rana
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (B.R.); (J.J.I.-H.); (P.S.R.)
- Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - James J. Driscoll
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (B.R.); (J.J.I.-H.); (P.S.R.)
- Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
26
|
Sutehall S, Malinsky F, Shurlock J, Wang G, Bosch A, Pitsiladis YP. Whole-Blood and Peripheral Mononuclear Cell Transcriptional Response to Prolonged Altitude Exposure in Well-Trained Runners. Clin J Sport Med 2023; 33:e135-e144. [PMID: 37656978 DOI: 10.1097/jsm.0000000000001046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 01/03/2022] [Indexed: 09/03/2023]
Abstract
BACKGROUND Recombinant human erythropoietin (rHuEpo) abuse by athletes threatens the integrity of sport. Due to the overlap in physiological response to rHuEpo and altitude exposure, it remains difficult to differentiate changes in hematological variables caused by rHuEpo or altitude, and therefore, other molecular methods to enhance anti-doping should be explored. OBJECTIVE To identify the hematological and transcriptomic response to prolonged altitude exposure typical of practices used by elite athletes. DESIGN Longitudinal study. SETTING University of Cape Town and Altitude Training Centre in Ethiopia. PARTICIPANTS AND INTERVENTION Fourteen well-trained athletes sojourned to an altitude training camp in Sululta, Ethiopia (∼2400-2500 m above sea level) for 27 days. Blood samples were taken before arrival, 24 hours, and 9, 16, and 24 days after arrival at altitude in addition to 24 hours and 6, 13, and 27 days upon return to sea level. MAIN OUTCOME MEASURES Blood samples were analyzed for hemoglobin concentration, hematocrit, and reticulocyte percentage. The transcriptomic response in whole blood and peripheral blood mononuclear cells (PBMC) were analyzed using gene expression microarrays. RESULTS A unique set of 29 and 10 genes were identified to be commonly expressed at every altitude time point in whole blood and PBMC, respectively. There were no genes identified upon return to sea level in whole blood, and only one gene within PBMC. CONCLUSIONS The current study has identified a series of unique genes that can now be integrated with genes previously validated for rHuEpo abuse, thereby enabling the differentiation of rHuEpo from altitude exposure.
Collapse
Affiliation(s)
- Shaun Sutehall
- Division of Physiological Sciences, University of Cape Town, Cape Town, South Africa
| | - Fernanda Malinsky
- Centre for Stress and Age-Related Disease, University of Brighton, Brighton, United Kingdom
| | | | - Guan Wang
- School of Sport and Health Sciences, University of Brighton, Brighton, United Kingdom
| | - Andrew Bosch
- Centre for Exercise Sciences and Sports Medicine, FIMS Collaborating Centre of Sports Medicine, Rome, Italy
| | - Yannis P Pitsiladis
- Centre for Stress and Age-Related Disease, University of Brighton, Brighton, United Kingdom
- Centre for Exercise Sciences and Sports Medicine, FIMS Collaborating Centre of Sports Medicine, Rome, Italy
- European Federation of Sports Medicine Associations (EFSMA), Lausanne, Switzerland; and
- International Federation of Sports Medicine (FIMS), Lausanne, Switzerland
| |
Collapse
|
27
|
Troise D, Infante B, Mercuri S, Netti GS, Ranieri E, Gesualdo L, Stallone G, Pontrelli P. Hypoxic State of Cells and Immunosenescence: A Focus on the Role of the HIF Signaling Pathway. Biomedicines 2023; 11:2163. [PMID: 37626660 PMCID: PMC10452839 DOI: 10.3390/biomedicines11082163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Hypoxia activates hypoxia-related signaling pathways controlled by hypoxia-inducible factors (HIFs). HIFs represent a quick and effective detection system involved in the cellular response to insufficient oxygen concentration. Activation of HIF signaling pathways is involved in improving the oxygen supply, promoting cell survival through anaerobic ATP generation, and adapting energy metabolism to meet cell demands. Hypoxia can also contribute to the development of the aging process, leading to aging-related degenerative diseases; among these, the aging of the immune system under hypoxic conditions can play a role in many different immune-mediated diseases. Thus, in this review we aim to discuss the role of HIF signaling pathways following cellular hypoxia and their effects on the mechanisms driving immune system senescence.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| |
Collapse
|
28
|
Akinsulie OC, Shahzad S, Ogunleye SC, Oladapo IP, Joshi M, Ugwu CE, Gbadegoye JO, Hassan FO, Adeleke R, Afolabi Akande Q, Adesola RO. Crosstalk between hypoxic cellular micro-environment and the immune system: a potential therapeutic target for infectious diseases. Front Immunol 2023; 14:1224102. [PMID: 37600803 PMCID: PMC10434535 DOI: 10.3389/fimmu.2023.1224102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
There are overwhelming reports on the promotional effect of hypoxia on the malignant behavior of various forms of cancer cells. This has been proposed and tested exhaustively in the light of cancer immunotherapy. However, there could be more interesting functions of a hypoxic cellular micro-environment than malignancy. There is a highly intricate crosstalk between hypoxia inducible factor (HIF), a transcriptional factor produced during hypoxia, and nuclear factor kappa B (NF-κB) which has been well characterized in various immune cell types. This important crosstalk shares common activating and inhibitory stimuli, regulators, and molecular targets. Impaired hydroxylase activity contributes to the activation of HIFs. Inflammatory ligands activate NF-κB activity, which leads to the expression of inflammatory and anti-apoptotic genes. The eventual sequelae of the interaction between these two molecular players in immune cells, either bolstering or abrogating functions, is largely cell-type dependent. Importantly, this holds promise for interesting therapeutic interventions against several infectious diseases, as some HIF agonists have helped prevent immune-related diseases. Hypoxia and inflammation are common features of infectious diseases. Here, we highlighted the role of this crosstalk in the light of functional immunity against infection and inflammation, with special focus on various innate and adaptive immune cells. Particularly, we discussed the bidirectional effects of this crosstalk in the regulation of immune responses by monocytes/macrophages, dendritic cells, neutrophils, B cells, and T cells. We believe an advanced understanding of the interplay between HIFs and NF-kB could reveal novel therapeutic targets for various infectious diseases with limited treatment options.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sammuel Shahzad
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Seto Charles Ogunleye
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Ifeoluwa Peace Oladapo
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Melina Joshi
- Center for Molecular Dynamics Nepal, Kathmandu, Nepal
| | - Charles Egede Ugwu
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
| | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Richard Adeleke
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
29
|
Li QQ, Zhang J, Wang HY, Niu SF, Wu RX, Tang BG, Wang QH, Liang ZB, Liang YS. Transcriptomic Response of the Liver Tissue in Trachinotus ovatus to Acute Heat Stress. Animals (Basel) 2023; 13:2053. [PMID: 37443851 DOI: 10.3390/ani13132053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Trachinotus ovatus is a major economically important cultured marine fish in the South China Sea. However, extreme weather and increased culture density result in uncontrollable problems, such as increases in water temperature and a decline in dissolved oxygen (DO), hindering the high-quality development of aquaculture. In this study, liver transcriptional profiles of T. ovatus were investigated under acute high-temperature stress (31 °C and 34 °C) and normal water temperature (27 °C) using RNA sequencing (RNA-Seq) technology. Differential expression analysis and STEM analysis showed that 1347 differentially expressed genes (DEGs) and four significant profiles (profiles 0, 3, 4, and 7) were screened, respectively. Of these DEGs, some genes involved in heat shock protein (HSPs), hypoxic adaptation, and glycolysis were up-regulated, while some genes involved in the ubiquitin-proteasome system (UPS) and fatty acid metabolism were down-regulated. Our results suggest that protein dynamic balance and function, hypoxia adaptation, and energy metabolism transformation are crucial in response to acute high-temperature stress. Our findings contribute to understanding the molecular response mechanism of T. ovatus under acute heat stress, which may provide some reference for studying the molecular mechanisms of other fish in response to heat stress.
Collapse
Affiliation(s)
- Qian-Qian Li
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Jing Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Hong-Yang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Su-Fang Niu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Ren-Xie Wu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Bao-Gui Tang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524025, China
| | - Qing-Hua Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhen-Bang Liang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yan-Shan Liang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
30
|
Jia Y, Wang F, Gao Y, Qin H, Guan C. Hypoxia stress induces hepatic antioxidant activity and apoptosis, but stimulates immune response and immune-related gene expression in black rockfish Sebastes schlegelii. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 258:106502. [PMID: 36965427 DOI: 10.1016/j.aquatox.2023.106502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/07/2022] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Dissolved oxygen concentrations both in the open ocean and coast have been declining due to the interaction of global climate change and human activity. Fish have evolved different adaptative strategies to cope with possibly damage induced by hypoxic environments. Black rockfish as important economic fish widely reared in the offshore sea cage, whereas related physiological response subject to hypoxia stress remained unclear. In this study, hepatic anti-oxidant enzymes (superoxide dismutase [SOD], catalase [CAT], glutathione peroxidase [GSH-Px]), aminotransferase (AST) and alanine aminotransferase (ALT) activities, lipid peroxidation (LPO), malondialdehyde (MDA) and glutathione (GSH) content, immunological parameters and the expression of apoptosis (bax, bcl2, p53, caspase3, xiap) and immune-related genes (c3, il-1β, ccl25, saa, hap, isg15) of black rockfish were determined during hypoxia and reoxygenation to illustrate the underlying defense response mechanisms. Results showed that hypoxia stress remarkably increased hepatic LPO and MDA content, AST and ALT activity and proportion of pyknotic nucleus. Hepatic SOD, CAT and GSH-Px activity manifested similar results, whereas GSH levels significantly decreased under hypoxia stress. The apoptosis rate of hepatocyte increased during hypoxia stress and reoxygenation. Meanwhile, p53, caspase3, bax and xiap mRNAs and bax/bcl2 rations were significantly up-regulated under hypoxia stress. However, bcl2 mRNA was significantly down-regulated. Interestingly, hypoxia stress significantly increased NBT-positive cell percent, phagocytic index, respiratory burst and ACH50 activity, and lysozyme activity. The mRNA levels of c3, ilβ, ccl25, saa, hap and isg15 were significantly up-regulated in the liver, spleen and head-kidney under hypoxia stress. The above parameters recovered to normal status after reoxygenation for 24 h Thus, hypoxia stress impairs hepatic antioxidant capacity, induces oxidative damage and apoptosis via the xiap-p53-bax-bcl2 and the caspase-dependent pathways, but enhances host immunity by regulating nonspecific immune indices and related genes expression to maintain homeostasis in black rockfish. These findings will help fully understand the hypoxia tolerance mechanisms of black rockfish and provide more data for offshore open ocean farming.
Collapse
Affiliation(s)
- Yudong Jia
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| | - Fenglin Wang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Yuntao Gao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Hongyu Qin
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Changtao Guan
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| |
Collapse
|
31
|
Liu N, Dong J, Li L, Liu F. Osteoimmune Interactions and Therapeutic Potential of Macrophage-Derived Small Extracellular Vesicles in Bone-Related Diseases. Int J Nanomedicine 2023; 18:2163-2180. [PMID: 37131544 PMCID: PMC10149074 DOI: 10.2147/ijn.s403192] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/19/2023] [Indexed: 05/04/2023] Open
Abstract
Due to the aging of the global population, the burden of bone-related diseases has increased sharply. Macrophage, as indispensable components of both innate immune responses and adaptive immunity, plays a considerable role in maintaining bone homeostasis and promoting bone establishment. Small extracellular vesicles (sEVs) have attracted increasing attention because they participate in cell cross-talk in pathological environments and can serve as drug delivery systems. In recent years, an increasing number of studies have expanded our knowledge about the effects of macrophage-derived sEVs (M-sEVs) in bone diseases via different forms of polarization and their biological functions. In this review, we comprehensively describe on the application and mechanisms of M-sEVs in various bone diseases and drug delivery, which may provide new perspectives for treating and diagnosing human bone disorders, especially osteoporosis, arthritis, osteolysis, and bone defects.
Collapse
Affiliation(s)
- Nan Liu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jinlei Dong
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Lianxin Li
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Fanxiao Liu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
32
|
Yao J, Liu J, He Y, Liu L, Xu Z, Lin X, Liu N, Kai G. Systems pharmacology reveals the mechanism of Astragaloside IV in improving immune activity on cyclophosphamide-induced immunosuppressed mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116533. [PMID: 37100262 DOI: 10.1016/j.jep.2023.116533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/20/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myelosuppression, also known as bone marrow suppression (BMS), is a pathological phenomenon of the decrease in the production of blood cells and further lead to immune homeostasis disorder. Astragalus mongholicus Bunge (AM, checked with The World Flora Online, http://www.worldfloraonline.org, updated on January 30, 2023) is a traditional Chinese medicine with efficacy of tonifying Qi and strengthening body immunity in thousands of years of clinical practice in China. Astragaloside IV (AS-IV) is a major active ingredient of AM, which plays an important role in regulating immune system through different ways. AIM OF THE STUDY This study was aimed to investigate the protective effect and mechanism of AS-IV on macrophages in vitro and cyclophosphamide (CTX)-induced immunosuppressive mice in vivo, and to provide experimental basis for the prevention and treatment of AS-IV in myelosuppression. MATERIALS AND METHODS Based on network pharmacology and molecular docking technology, the core targets and signaling pathways of saponins of AM against myelosuppression were screened. And then, the immunoregulatory effect of AS-IV on RAW264.7 cells was investigated by cellular immune activity and cellular secretion analysis in vitro. In this way, the effects of AS-IV on the main potential targets of HIF-1α/NF-κB signaling pathway were analyzed by qRT-PCR and Western blot methods. Furthermore, comprehensive analysis of the effects of AS-IV against CTX-induced mice were conducted on the basis of immune organs indices analysis, histopathological analysis, hematological analysis, natural killer cell activity analysis and spleen lymphocyte transformation activity analysis. In order to further verify the relationship between active ingredients and action targets, drug inhibitor experiments were finally conducted. RESULTS AS-IV, as a potential anti-myelosuppressive compound, was screened by systematic pharmacological methods to act on target genes including HIF1A and RELA together with the HIF-1α/NF-κB signaling pathway. Further studies by molecular docking technology showed that AS-IV had good binding activity with HIF1A, RELA, TNF, IL6, IL1B and other core targets. Besides, cellular and animal experiments validation results showed that AS-IV could enhance the migration and phagocytosis of RAW264.7 cells, and protect the immune organs such as spleen and thymus together with bone tissues from damage. By this means, immune cell function including spleen natural killer cell and lymphocyte transformation activity were also enhanced. In addition, white blood cells, red blood cells, hemoglobin, platelets and bone marrow cells were also significantly improved in the suppressed bone marrow microenvironment (BMM). In kinetic experiments, the secretion of cytokines such as TNF-α, IL-6 and IL-1β were increased, and IL-10, TGF-β1 were decreased. The key regulatory proteins such as HIF-1α, NF-κB, PHD3 in HIF-1α/NF-κB signaling pathway were also regulated in the results of upregulated expression of HIF-1α, p-NF-κB p65 and PHD3 at the protein or mRNA level. Finally, the inhibition experiment results suggested that AS-IV could significantly improve protein response in immunity and inflammation such as HIF-1α, NF-κB and PHD3. CONCLUSION AS-IV could significantly relieve CTX-induced immunosuppressive and might improve the immune activity of macrophages by activating HIF-1α/NF-κB signaling pathway, and provide a reliable basis for the clinical application of AS-IV as a potentially valuable regulator of BMM.
Collapse
Affiliation(s)
- Jiaxiong Yao
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Junqiu Liu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yining He
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Lin Liu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Zonghui Xu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Xianming Lin
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Na Liu
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Guoyin Kai
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, Jinhua Academy, School of Pharmaceutical Sciences, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
33
|
Nourkami-Tutdibi N, Küllmer J, Dietrich S, Monz D, Zemlin M, Tutdibi E. Serum vascular endothelial growth factor is a potential biomarker for acute mountain sickness. Front Physiol 2023; 14:1083808. [PMID: 37064896 PMCID: PMC10098311 DOI: 10.3389/fphys.2023.1083808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
Background: Acute mountain sickness (AMS) is the most common disease caused by hypobaric hypoxia (HH) in high-altitude (HA) associated with high mortality when progressing to high-altitude pulmonary edema (HAPE) and/or high-altitude cerebral edema (HACE). There is evidence for a role of pro- and anti-inflammatory cytokines in development of AMS, but biological pathways and molecular mechanisms underlying AMS remain elusive. We aimed to measure changes in blood cytokine levels and their possible association with the development of AMS.Method: 15 healthy mountaineers were included into this prospective clinical trial. All participants underwent baseline normoxic testing with venous EDTA blood sampling at the Bangor University in United Kingdom (69 m). The participants started from Beni at an altitude of 869 m and trekked same routes in four groups the Dhaulagiri circuit in the Nepali Himalaya. Trekking a 14-day route, the mountaineers reached the final HA of 5,050 m at the Hidden Valley Base Camp (HVBC). Venous EDTA blood sampling was performed after active ascent to HA the following morning after arrival at 5,050 m (HVBC). A panel of 21 cytokines, chemokines and growth factors were assessed using Luminex system (IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p40, IL-1ra, sIL-2Rα, IFN-γ, TNF-α, MCP-1, MIP-1α, MIP-1β, IP-10, G-CSF, GM-CSF, EGF, FGF-2, VEGF, and TGF-β1).Results: There was a significant main effect for the gradual ascent from sea-level (SL) to HA on nearly all cytokines. Serum levels for TNF-α, sIL-2Rα, G-CSF, VEGF, EGF, TGF-β1, IL-8, MCP-1, MIP-1β, and IP-10 were significantly increased at HA compared to SL, whereas levels for IFN-γ and MIP-1α were significantly decreased. Serum VEGF was higher in AMS susceptible versus AMS resistant subjects (p < 0.027, main effect of AMS) and increased after ascent to HA in both AMS groups (p < 0.011, main effect of HA). Serum VEGF increased more from SL values in the AMS susceptible group than in the AMS resistant group (p < 0.049, interaction effect).Conclusion: Cytokine concentrations are significantly altered in HA. Within short interval after ascent, cytokine concentrations in HH normalize to values at SL. VEGF is significantly increased in mountaineers suffering from AMS, indicating its potential role as a biomarker for AMS.
Collapse
|
34
|
David BT, Curtin JJ, Brown JL, Scorpio K, Kandaswamy V, Coutts DJC, Vivinetto A, Bianchimano P, Karuppagounder SS, Metcalfe M, Cave JW, Hill CE. Temporary induction of hypoxic adaptations by preconditioning fails to enhance Schwann cell transplant survival after spinal cord injury. Glia 2023; 71:648-666. [PMID: 36565279 PMCID: PMC11848738 DOI: 10.1002/glia.24302] [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: 04/19/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 12/25/2022]
Abstract
Hypoxic preconditioning is protective in multiple models of injury and disease, but whether it is beneficial for cells transplanted into sites of spinal cord injury (SCI) is largely unexplored. In this study, we analyzed whether hypoxia-related preconditioning protected Schwann cells (SCs) transplanted into the contused thoracic rat spinal cord. Hypoxic preconditioning was induced in SCs prior to transplantation by exposure to either low oxygen (1% O2 ) or pharmacological agents (deferoxamine or adaptaquin). All preconditioning approaches induced hypoxic adaptations, including increased expression of HIF-1α and its target genes. These adaptations, however, were transient and resolved within 24 h of transplantation. Pharmacological preconditioning attenuated spinal cord oxidative stress and enhanced transplant vascularization, but it did not improve either transplanted cell survival or recovery of sensory or motor function. Together, these experiments show that hypoxia-related preconditioning is ineffective at augmenting either cell survival or the functional outcomes of SC-SCI transplants. They also reveal that the benefits of hypoxia-related adaptations induced by preconditioning for cell transplant therapies are not universal.
Collapse
Affiliation(s)
- Brian T. David
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Jessica J. Curtin
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Jennifer L. Brown
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Kerri Scorpio
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Veena Kandaswamy
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - David J. C. Coutts
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Ana Vivinetto
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Paola Bianchimano
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Saravanan S. Karuppagounder
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Mariajose Metcalfe
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - John W. Cave
- InVitro Cell Research, LLC, Englewood, NJ, United States
| | - Caitlin E. Hill
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
- Neural Stem Cell Institute, Rensselaer, NY, United States
| |
Collapse
|
35
|
Goodin DA, Frieboes HB. Evaluation of innate and adaptive immune system interactions in the tumor microenvironment via a 3D continuum model. J Theor Biol 2023; 559:111383. [PMID: 36539112 DOI: 10.1016/j.jtbi.2022.111383] [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: 05/06/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Immune cells in the tumor microenvironment (TME) are known to affect tumor growth, vascularization, and extracellular matrix (ECM) deposition. Marked interest in system-scale analysis of immune species interactions within the TME has encouraged progress in modeling tumor-immune interactions in silico. Due to the computational cost of simulating these intricate interactions, models have typically been constrained to representing a limited number of immune species. To expand the capability for system-scale analysis, this study develops a three-dimensional continuum mixture model of tumor-immune interactions to simulate multiple immune species in the TME. Building upon a recent distributed computing implementation that enables efficient solution of such mixture models, major immune species including monocytes, macrophages, natural killer cells, dendritic cells, neutrophils, myeloid-derived suppressor cells (MDSC), cytotoxic, helper, regulatory T-cells, and effector and regulatory B-cells and their interactions are represented in this novel implementation. Immune species extravasate from blood vasculature, undergo chemotaxis toward regions of high chemokine concentration, and influence the TME in proportion to locally defined levels of stimulation. The immune species contribute to the production of angiogenic and tumor growth factors, promotion of myofibroblast deposition of ECM, upregulation of angiogenesis, and elimination of living and dead tumor species. The results show that this modeling approach offers the capability for quantitative insight into the modulation of tumor growth by diverse immune-tumor interactions and immune-driven TME effects. In particular, MDSC-mediated effects on tumor-associated immune species' activation levels, volume fraction, and influence on the TME are explored. Longer term, linking of the model parameters to particular patient tumor information could simulate cancer-specific immune responses and move toward a more comprehensive evaluation of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Dylan A Goodin
- Department of Bioengineering, University of Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, KY, USA; Center for Predictive Medicine, University of Louisville, KY, USA.
| |
Collapse
|
36
|
Matthiesen S, Christiansen B, Jahnke R, Zaeck LM, Karger A, Finke S, Franzke K, Knittler MR. TGF-β/IFN-γ Antagonism in Subversion and Self-Defense of Phase II Coxiella burnetii -Infected Dendritic Cells. Infect Immun 2023; 91:e0032322. [PMID: 36688662 PMCID: PMC9933720 DOI: 10.1128/iai.00323-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) belong to the first line of innate defense and come into early contact with invading pathogens, including the zoonotic bacterium Coxiella burnetii, the causative agent of Q fever. However, the pathogen-host cell interactions in C. burnetii-infected DCs, particularly the role of mechanisms of immune subversion beyond virulent phase I lipopolysaccharide (LPS), as well as the contribution of cellular self-defense strategies, are not understood. Using phase II Coxiella-infected DCs, we show that impairment of DC maturation and MHC I downregulation is caused by autocrine release and action of immunosuppressive transforming growth factor-β (TGF-β). Our study demonstrates that IFN-γ reverses TGF-β impairment of maturation/MHC I presentation in infected DCs and activates bacterial elimination, predominantly by inducing iNOS/NO. Induced NO synthesis strongly affects bacterial growth and infectivity. Moreover, our studies hint that Coxiella-infected DCs might be able to protect themselves from mitotoxic NO by switching from oxidative phosphorylation to glycolysis, thus ensuring survival in self-defense against C. burnetii. Our results provide new insights into DC subversion by Coxiella and the IFN-γ-mediated targeting of C. burnetii during early steps in the innate immune response.
Collapse
Affiliation(s)
- Svea Matthiesen
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Bahne Christiansen
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Rico Jahnke
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Kati Franzke
- Institute of Infectology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Michael R. Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| |
Collapse
|
37
|
Hypoxia and Intestinal Inflammation: Common Molecular Mechanisms and Signaling Pathways. Int J Mol Sci 2023; 24:ijms24032425. [PMID: 36768744 PMCID: PMC9917195 DOI: 10.3390/ijms24032425] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The gastrointestinal tract (GI) has a unique oxygenation profile. It should be noted that the state of hypoxia can be characteristic of both normal and pathological conditions. Hypoxia-inducible factors (HIF) play a key role in mediating the response to hypoxia, and they are tightly regulated by a group of enzymes called HIF prolyl hydroxylases (PHD). In this review, we discuss the involvement of inflammation hypoxia and signaling pathways in the pathogenesis of inflammatory bowel disease (IBD) and elaborate in detail on the role of HIF in multiple immune reactions during intestinal inflammation. We emphasize the critical influence of tissue microenvironment and highlight the existence of overlapping functions and immune responses mediated by the same molecular mechanisms. Finally, we also provide an update on the development of corresponding therapeutic approaches that would be useful for treatment or prophylaxis of inflammatory bowel disease.
Collapse
|
38
|
Peng C, Ye H, li Z, Duan X, Yang W, Yi Z. Multi-omics characterization of a scoring system to quantify hypoxia patterns in patients with head and neck squamous cell carcinoma. J Transl Med 2023; 21:15. [PMID: 36627705 PMCID: PMC9830846 DOI: 10.1186/s12967-022-03869-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The 5-year survival rate of patients with head and neck squamous cell carcinoma (HNSCC) remains < 50%. Hypoxia patterns are a hallmark of HNSCC that are associated with its occurrence and progression. However, the precise role of hypoxia during HNSCC, such as the relationship between hypoxia, tumor immune landscape and cell communication orchestration remains largely unknown. The current study integrated data from bulk and single-cell RNA sequencing analyses to define the relationship between hypoxia and HNSCC. METHODS A scoring system named the hypoxia score (HS) was constructed based on hypoxia-related genes (HRGs) expression. The predictive value of HS response for patient outcomes and different treatments was evaluated. Single-cell datasets and cell communication were utilized to rule out cell populations which hypoxia targeted on. RESULTS The survival outcomes, immune/Estimate scores, responses to targeted inhibitors, and chemotherapeutic, and immunotherapy responses were distinct between a high HS group and a low HS group (all P < 0.05). Single-cell datasets showed different distributions of HS in immune cell populations (P < 0.05). Furthermore, HLA-DPA1/CD4 axis was identified as a unique interaction between CD4 + T Conv and pDC cells. CONCLUSIONS Altogether, the quantification for hypoxia patterns is a potential biomarker for prognosis, individualized chemotherapeutic and immunotherapy strategies. The portrait of cell communication characteristics over the HNSCC ecosystem enhances the understanding of hypoxia patterns in HNSCC.
Collapse
Affiliation(s)
- Cong Peng
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Huiping Ye
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zhengyang li
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiaofeng Duan
- grid.459540.90000 0004 1791 4503Department of Oral and Maxillofacial Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Wen Yang
- grid.452244.1Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhuguang Yi
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
39
|
Dzhalilova DS, Kosyreva AM, Tsvetkov IS, Zolotova NA, Sentyabreva AV, Makarova OV. Morphofunctional Changes in the Thymus in Prepubertal Male Wistar Rats in LPS-Induced Systemic Inflammatory Response in Relation to Hypoxia Tolerance. Bull Exp Biol Med 2023; 174:385-390. [PMID: 36723748 DOI: 10.1007/s10517-023-05713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Indexed: 02/02/2023]
Abstract
The dynamics of morphofunctional changes in the thymus during the LPS-induced systemic inflammatory response was assessed in prepubertal male Wistar rats in relationship with the resistance to hypoxia. The systemic inflammatory response was modeled by intraperitoneal administration of E. coli O26:B6 LPS. In histological sections of the thymus, the relative number of thymic bodies and proliferative activity of cells were evaluated. The relative number of CD3+CD4+, CD3+CD8+, and CD4+CD8+ cells in the thymus was determined by flow cytometry. The content of HIF-1α and endotoxin was determined in the blood serum. The expression level of Nfkb mRNA was assessed in the liver. The most pronounced changes in the indicators of the functional state of the thymus were detected 3 and 6 h after LPS administration following the increase in the content of HIF-1α and endotoxin in blood serum and Nfkb mRNA expression in the liver. In the thymus, a decrease in the number of thymic bodies consisting of 3-5 epithelial cells and an increase in the number of bodies consisting of 5 or more cells was observed. In 24 h after LPS administration, the relative number of CD3+CD4+ and CD3+CD8+ cells in the thymus decreased. At the same time, the number of Ki-67+ cells in the subcapsular zone of the thymus increased 6 and 24 h after LPS administration. These data should be taken into account in the development of approaches to the treatment of infectious and inflammatory diseases in prepubertal children.
Collapse
Affiliation(s)
- D Sh Dzhalilova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia.
| | - A M Kosyreva
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - I S Tsvetkov
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - N A Zolotova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - A V Sentyabreva
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - O V Makarova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky National Research Centre of Surgery, Moscow, Russia
| |
Collapse
|
40
|
Mougel A, Méjean F, Tran T, Adimi Y, Galy-Fauroux I, Kaboré C, Mercier E, Urquia P, Terme M, Tartour E, Tanchot C. Synergistic effect of combining sunitinib with a peptide-based vaccine in cancer treatment after microenvironment remodeling. Oncoimmunology 2022; 11:2110218. [PMID: 35968405 PMCID: PMC9367646 DOI: 10.1080/2162402x.2022.2110218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Although it has proven difficult to demonstrate the clinical efficacy of therapeutic vaccination as a monotherapy in advanced cancers, its combination with an immunomodulatory treatment to reduce intra-tumor immunosuppression and improve vaccine efficacy is a very promising strategy. In this context, we are studying the combination of a vaccine composed of peptides of the tumor antigen survivin (SVX vaccine) with the anti-angiogenic agent sunitinib in a colorectal carcinoma model. To this end, we have been focusing on administration scheduling and have highlighted a therapeutic synergy between SVX vaccine and sunitinib when the vaccine was administered at the end of anti-angiogenic treatment. In this setting, a prolonged control of tumor growth associated with an important percentage of complete tumor regression was observed. Studying the remodeling induced by each therapy on the immunological and angiogenic tumor microenvironment over time we observed, during sunitinib treatment, a transient increase in polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and a decrease in NK cells in the tumor microenvironment. In contrast, after sunitinib treatment was stopped, a decrease in PMN-MDSC populations has been observed in the tumor, associated with an increase in NK cells, pericyte coverage of tumor vessels and CD8+ T cell population and functionality. In conclusion, sunitinib treatment results in the promotion of an immune-favorable tumor microenvironment that can guide the optimal sequence of vaccine and anti-angiogenic combination to reinforce their synergy.
Collapse
Affiliation(s)
- Alice Mougel
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Fanny Méjean
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Thi Tran
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Yasmine Adimi
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | | | | | - Erwan Mercier
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Pauline Urquia
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Magali Terme
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
| | - Eric Tartour
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France
- Department of Immunology, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | | |
Collapse
|
41
|
Abstract
Bacteria engulfed by phagocytic cells must resist oxidation damage and adapt to cellular hypoxia, but the mechanisms involved in this process are not completely elucidated. Recent work by Kim et al. in the Journal of Biological Chemistry investigated how the intracellular pathogen Salmonella enterica activates gene expression required to counteract oxidative damage. The authors show that this bacterium utilizes host oxidative molecules to activate regulatory proteins that enhance the production of effector molecules, counteracting the host weapon NADPH oxidase and inducing a protective response.
Collapse
|
42
|
da Silva-Ferreira S, Duarte-Oliveira C, Antunes D, Barbosa-Matos C, Mendes-Frias A, Torrado E, Costa S, Silvestre R, Cunha C, Carvalho A. Hypoxia inducible-factor 1 alpha regulates neutrophil recruitment during fungal-elicited granulomatous inflammation. Front Cell Infect Microbiol 2022; 12:1005839. [PMID: 36275017 PMCID: PMC9582458 DOI: 10.3389/fcimb.2022.1005839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Abstract
Chronic pulmonary aspergillosis (CPA) is a devastating disease with increasing prevalence worldwide. The characteristic granulomatous-like inflammation poses as the major setback to effective antifungal therapies by limiting drug access to fungi. These inflammatory lung structures are reported to be severely hypoxic; nevertheless, the underlying mechanisms whereby these processes contribute to fungal persistence remain largely unknown. Hypoxia-inducible factor 1 alpha (HIF-1α), besides being the major cellular response regulator to hypoxia, is a known central immune modulator. Here, we used a model of Aspergillus fumigatus airway infection in myeloid-restricted HIF-1α knock-out (mHif1α-/-) mice to replicate the complex structures resembling fungal granulomas and evaluate the contribution of HIF-1α to antifungal immunity and disease development. We found that fungal-elicited granulomas in mHif1α-/- mice had significantly smaller areas, along with extensive hyphal growth and increased lung fungal burden. This phenotype was associated with defective neutrophil recruitment and an increased neutrophil death, therefore highlighting a central role for HIF-1α-mediated regulation of neutrophil function in the pathogenesis of chronic fungal infection. These results hold the promise of an improved capacity to manage the progression of chronic fungal disease and open new avenues for additional therapeutic targets and niches of intervention.
Collapse
Affiliation(s)
- Sara da Silva-Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Cláudio Duarte-Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Daniela Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Catarina Barbosa-Matos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Ana Mendes-Frias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Egídio Torrado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Sandra Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s– PT Government Associate Laboratory, Guimarães/Braga, Portugal
- *Correspondence: Agostinho Carvalho,
| |
Collapse
|
43
|
Xiao Y, Ding Y, Zhuang J, Sun R, Sun H, Bai L. Osteoimmunomodulation role of exosomes derived from immune cells on osseointegration. Front Bioeng Biotechnol 2022; 10:989537. [PMID: 36061437 PMCID: PMC9437288 DOI: 10.3389/fbioe.2022.989537] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022] Open
Abstract
Despite the high success rate of biomedical implants adopted clinically, implant failures caused by aseptic loosening still raise the risk of secondary surgery and a substantial economic burden to patients. Improving the stable combination between the implant and the host bone tissue, achieving fast and high-quality osseointegration can effectively reduce the probability of aseptic loosening. Accumulating studies have shown that the osteoimmunomodulation mediated by immune cells mainly dominated by macrophages plays a pivotal role in osseointegration by releasing active factors to improve the inflammatory microenvironment. However, the mechanism by which osteoimmunomodulation mediates osseointegration remains unclear. Recent studies have revealed that exosomes released by macrophages play a central role in mediating osteoimmunomodulation. The exosomes can be internalized by various cells participating in de novo bone formation, such as endothelial cells and osteoblasts, to intervene in the osseointegration robustly. Therefore, macrophage-derived exosomes with multifunctionality are expected to significantly improve the osseointegration microenvironment, which is promising in reducing the occurrence of aseptic loosening. Based on this, this review summarizes recent studies on the effects of exosomes derived from the immune cells on osseointegration, aiming to provide a theoretical foundation for improving the clinical success rate of biomedical implants and achieving high-quality and high-efficiency osseointegration.
Collapse
Affiliation(s)
- Yunchao Xiao
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, China
- Nanotechnology Research Institute, Jiaxing University, Jiaxing, China
| | - Yanshu Ding
- Engineering Research Center for Biomedical Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Jingwen Zhuang
- Engineering Research Center for Biomedical Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Ruoyue Sun
- Engineering Research Center for Biomedical Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Hui Sun
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
44
|
Zhang J, Wu X, Ma J, Long K, Sun J, Li M, Ge L. Hypoxia and hypoxia-inducible factor signals regulate the development, metabolism, and function of B cells. Front Immunol 2022; 13:967576. [PMID: 36045669 PMCID: PMC9421003 DOI: 10.3389/fimmu.2022.967576] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/26/2022] [Indexed: 11/28/2022] Open
Abstract
Hypoxia is a common hallmark of healthy tissues in physiological states or chronically inflamed tissues in pathological states. Mammalian cells sense and adapt to hypoxia mainly through hypoxia-inducible factor (HIF) signaling. Many studies have shown that hypoxia and HIF signaling play an important regulatory role in development and function of innate immune cells and T cells, but their role in B cell biology is still controversial. B cells experience a complex life cycle (including hematopoietic stem cells, pro-B cells, pre-B cells, immature B cells, mature naïve B cells, activated B cells, plasma cells, and memory B cells), and the partial pressure of oxygen (PO2) in the corresponding developmental niche of stage-specific B cells is highly dynamic, which suggests that hypoxia and HIF signaling may play an indispensable role in B cell biology. Based on the fact that hypoxia niches exist in the B cell life cycle, this review focuses on recent discoveries about how hypoxia and HIF signaling regulate the development, metabolism, and function of B cells, to facilitate a deep understanding of the role of hypoxia in B cell-mediated adaptive immunity and to provide novel strategies for vaccine adjuvant research and the treatment of immunity-related or infectious diseases.
Collapse
Affiliation(s)
- Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
- Chongqing Camab Biotech Ltd., Chongqing, China
| | - Xiaoqian Wu
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Jideng Ma
- Chongqing Academy of Animal Sciences, Chongqing, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Keren Long
- Chongqing Academy of Animal Sciences, Chongqing, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Mingzhou Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
- Chongqing Camab Biotech Ltd., Chongqing, China
| |
Collapse
|
45
|
Lu Y, Xu L, Lin X, Qi X, Xia L, Wang Y. Analysis of the Effect of Fast Recovery Surgery Concept on Perioperative Nursing Care of Patients with Radical Resection of Cervical Cancer and Its Influence on Psychological Status. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2023159. [PMID: 35983010 PMCID: PMC9381187 DOI: 10.1155/2022/2023159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
Objective To explore the applied effect of fast-track surgery concept in the perioperative nursing of patients undergoing radical cervical cancer surgery and its influence on mental state. Methods The clinical data of 110 patients undergoing radical cervical cancer surgery in our hospital from May 2015 to May 2017 were retrospectively analyzed, and they were randomly divided into a research group (n = 55) and a reference group (n = 55). The reference group received routine clinical nursing, and the research group received fast-track surgical nursing. Then, we compared their nursing effect and influence on the mental state of patients in the two groups. Results The Karnofsky performance status (KPS) scores of patients in the two groups after intervention were significantly lower than those before the intervention (P < 0.001), and after intervention, patients in the research group had significantly lower KPS scores as compared to patients in the reference group (P < 0.001). There was a significant decrease in the self-rating anxiety scale (SAS) scores and self-rating depression scale (SDS) scores of patients in the two groups after intervention in comparison with before intervention (P < 0.001), and patients in the research group after intervention had significantly lower SAS and SDS scores compared to the reference group (P < 0.001). Patients in the research group spent short time on expelling gas, eating, and getting out of bed as compared to the reference group (P < 0.001); after intervention, compared to patients in the reference group, patients in the research group had significantly higher scores in cognitive function, emotional function, social function, and physical function (P < 0.05). There was no significant difference in IgA, IgM, and IgG levels before intervention (P > 0.05). After intervention, the IgA, IgM, and IgG levels of patients in the two groups were all lower than before intervention, and patients in the research group had significantly higher IgA, IgM, and IgG levels as compared to the reference group (P < 0.001); the complication rate of patients in the research group was significantly lower than that in the reference group (P < 0.05). Conclusion The fast-track surgery concept effectively helps improve the negative emotion of patients, shorten recovery time, improve quality of life, and reduce the impact on immune function in the radical cervical cancer surgery, and it is worthy of promotion and application, with a high safety.
Collapse
Affiliation(s)
- Yun Lu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Luxi Xu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoyu Lin
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xueling Qi
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ling Xia
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
46
|
Construction of a hypoxia-derived gene model to predict the prognosis and therapeutic response of head and neck squamous cell carcinoma. Sci Rep 2022; 12:13538. [PMID: 35945448 PMCID: PMC9363468 DOI: 10.1038/s41598-022-17898-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) ranks as the sixth most common cancer worldwide and has a poor prognosis in the advanced stage. Increasing evidence has shown that hypoxia contributes to genetic alterations that have essential effects on the occurrence and progression of cancers. However, the exact roles hypoxia-related genes play in HNSCC remain unclear. In this study, we downloaded the mRNA expression profiles and clinical data of patients with HNSCC from The Cancer Genome Atlas and Gene Expression Omnibus. Two molecular subtypes were identified based on prognostic hypoxia-related genes using the ConsensusClusterPlus method. ESTIMATE was used to calculate the immune score of each patient. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology were used for functional annotation. A prognostic risk model was generated by Cox regression and least absolute shrinkage and selection operator analysis. We identified two distinct molecular subtypes, cluster 1 and cluster 2, based on 200 hypoxia-related genes. Additionally, we identified three hypoxia-immune subgroups (hypoxia-high/immune-low, hypoxia-low/immune-high, and mixed subgroups). The hypoxia-high/immune-low group had the worst prognosis, while the hypoxia-low/immune-high group had the best prognosis. Patients in the hypoxia-low/immune-high group were more sensitive to anti-PD-L1 treatment and chemotherapy than those in the hypoxia-high/immune-low group. Furthermore, we constructed a prognostic risk model based on the differentially expressed genes between the hypoxia-immune subgroups. The survival analysis and time-dependent ROC analysis results demonstrated the good performance of the established 7-gene signature for predicting HNSCC prognosis. In conclusions, the constructed hypoxia-related model might serve as a promising biomarker for the diagnosis and prognosis of HNSCC, and it could predict immunotherapy and chemotherapy efficacy in HNSCC.
Collapse
|
47
|
Li W, Yuan P, Liu W, Xiao L, Xu C, Mo Q, Xu S, He Y, Jiang D, Wang X. Hypoxia–Immune-Related Gene SLC19A1 Serves as a Potential Biomarker for Prognosis in Multiple Myeloma. Front Immunol 2022; 13:843369. [PMID: 35958555 PMCID: PMC9358019 DOI: 10.3389/fimmu.2022.843369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background Multiple myeloma (MM) remains an incurable malignant tumor of plasma cells. Increasing evidence has reported that hypoxia and immune status contribute to the progression of MM. In this research, the prognostic value of the hypoxia–immune-related gene SLC19A1 in MM was evaluated by bioinformatics analysis. Method RNA-sequencing (RNA-seq) data along with clinical information on MM were downloaded from the Gene Expression Omnibus (GEO) database. Consistent clustering analysis and ESTIMATE algorithms were performed to establish the MM sample subgroups related to hypoxia and immune status, respectively, based on the GSE24080 dataset. The differentially expressed analysis was performed to identify the hypoxia–immune-related genes. Subsequently, a hypoxia–immune-gene risk signature for MM patients was constructed by univariate and multivariate Cox regression analyses, which was also verified in the GSE4581 dataset. Furthermore, the mRNA expression of SLC19A1 was determined using qRT-PCR in 19 MM patients, and the correlations between the genetic expression of SLC19A1 and clinical features were further analyzed. Result A total of 47 genes were identified as hypoxia–immune-related genes for MM. Among these genes, SLC19A1 was screened to construct a risk score model that had better predictive power for MM. The constructed prognostic signature based on SLC19A1 was verified in the GSE4581 dataset. All independent prognostic factors (age, β2-microglobulin, LDH, albumin, MRI, and gene risk score) were used to develop a nomogram that showed a better performance for predicting the survival probability of MM patients for 1–5 years. Furthermore, SLC19A1 was highly expressed in newly diagnosed and relapsed MM patients, and high expression of SLC19A1 is correlated with higher bone marrow aspiration plasma cells and β2-microglobulin levels in MM patients. Conclusion In conclusion, our results suggest that SLC19A1 is aberrantly expressed in MM and highly expressed SLC19A1 might be a biomarker correlated with inferior prognosis. More importantly, we identified SLC19A1 as a hypoxia–immune-related gene in MM. Future functional and mechanistic studies will further clarify the roles of SLC19A1 in MM.
Collapse
Affiliation(s)
- Wenjin Li
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Peng Yuan
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Weiqin Liu
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Lichan Xiao
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Chun Xu
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Qiuyu Mo
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
- Department of Hematology, Second Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Shujuan Xu
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yuchan He
- Department of Hematology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Duanfeng Jiang
- Department of Hematology, Second Affiliated Hospital of Hainan Medical College, Haikou, China
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaotao Wang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Xiaotao Wang,
| |
Collapse
|
48
|
Shen R, Peng L, Zhou W, Wang D, Jiang Q, Ji J, Hu F, Yuan H. Anti-angiogenic nano-delivery system promotes tumor vascular normalizing and micro-environment reprogramming in solid tumor. J Control Release 2022; 349:550-564. [PMID: 35841997 DOI: 10.1016/j.jconrel.2022.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Aberrant tumor vasculature leads to the malignant tumor microenvironment (TME) for tumor progression. Research has found temporary tumor vascular normalization after treated with low-dose anti-angiogenic agents, however, has paid little attention to prolonging the normalization window and its further influence on tumor tissue. Based on the dose- and time-dependent effect of anti-angiogenic agents, we developed V@LDL NPs, a nano-delivery system sustainedly releasing Vandetanib, an anti-VEGFR2 inhibitor, to control the dose of drug to the normalizing level, and prove its stable tumor vascular normalizing effect in 4 T1 breast cancer model. Furthermore, long-term normalized vasculature could improve tumor perfusion, then provide a circulation to reverse abnormalities in TME, such as hypoxia and heterogeneity, and also inhibit tumor progression. Our findings demonstrate that stable tumor vascular normalization could be a considerable strategy for long-term change to remodel TME and probably result in a therapeutic benefit to anti-cancer treatment, which could be achieved by anti-angiogenic nano-delivery system.
Collapse
Affiliation(s)
- Ruoyu Shen
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Lijun Peng
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Wentao Zhou
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Ding Wang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Jian Ji
- Department of Polymer Science and Engineering, Zhejiang University, 38 Zhe Da Road, Hangzhou 310027, Zhejiang Province, People's Republic of China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| |
Collapse
|
49
|
Nedunchezhiyan U, Varughese I, Sun AR, Wu X, Crawford R, Prasadam I. Obesity, Inflammation, and Immune System in Osteoarthritis. Front Immunol 2022; 13:907750. [PMID: 35860250 PMCID: PMC9289681 DOI: 10.3389/fimmu.2022.907750] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity remains the most important risk factor for the incidence and progression of osteoarthritis (OA). The leading cause of OA was believed to be overloading the joints due to excess weight which in turn leads to the destruction of articular cartilage. However, recent studies have proved otherwise, various other factors like adipose deposition, insulin resistance, and especially the improper coordination of innate and adaptive immune responses may lead to the initiation and progression of obesity-associated OA. It is becoming increasingly evident that multiple inflammatory cells are recruited into the synovial joint that serves an important role in pathological changes in the synovial joint. Polarization of macrophages and macrophage-produced mediators are extensively studied and linked to the inflammatory and destructive responses in the OA synovium and cartilage. However, the role of other major innate immune cells such as neutrophils, eosinophils, and dendritic cells in the pathogenesis of OA has not been fully evaluated. Although cells of the adaptive immune system contribute to the pathogenesis of obesity-induced OA is still under exploration, a quantity of literature indicates OA synovium has an enriched population of T cells and B cells compared with healthy control. The interplay between a variety of immune cells and other cells that reside in the articular joints may constitute a vicious cycle, leading to pathological changes of the articular joint in obese individuals. This review addresses obesity and the role of all the immune cells that are involved in OA and summarised animal studies and human trials and knowledge gaps between the studies have been highlighted. The review also touches base on the interventions currently in clinical trials, different stages of the testing, and their shortcomings are also discussed to understand the future direction which could help in understanding the multifactorial aspects of OA where inflammation has a significant function.
Collapse
Affiliation(s)
- Udhaya Nedunchezhiyan
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ibin Varughese
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia RuJia Sun
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiaoxin Wu
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ross Crawford
- Orthopedic Department, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- *Correspondence: Indira Prasadam,
| |
Collapse
|
50
|
Hu J, Li X, Yang L, Li H. Hypoxia, a key factor in the immune microenvironment. Biomed Pharmacother 2022; 151:113068. [PMID: 35676780 DOI: 10.1016/j.biopha.2022.113068] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
The physical and chemical pressures in the tumor microenvironment (TME) play an important role in tumor development by regulating stromal elements, including immune cells. Hypoxia can induce a cascade of events in tumor initiation and development via immune regulation. As a dangerous factor, hypoxia activates multiple signaling pathways to reshape the immune microenvironment, leading to immunosuppression. Consequently, targeting hypoxia in the TME is a potential strategy to prevent immune escape and inhibit malignant tumor progression. In this review, we summarized the role of hypoxia-induced factors in the tumor immune escape process and provide a novel pathway to restrain tumor progression and development.
Collapse
Affiliation(s)
- Jingyao Hu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| |
Collapse
|