1
|
Van Kerkhove O, Verfaillie S, Maes B, Cuppens K. The Adenosinergic Pathway in Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:3142. [PMID: 39335114 PMCID: PMC11430550 DOI: 10.3390/cancers16183142] [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: 07/23/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-(L)1 and CTLA-4 have revolutionized the systemic treatment of non-small cell lung cancer (NSCLC), achieving impressive results. However, long-term clinical benefits are only seen in a minority of patients. Extensive research is being conducted on novel potential immune checkpoints and the mechanisms underlying ICI resistance. The tumor microenvironment (TME) plays a critical role in modulating the immune response and influencing the efficacy of ICIs. The adenosinergic pathway and extracellular adenosine (eADO) are potential targets to improve the response to ICIs in NSCLC patients. First, this review delves into the adenosinergic pathway and the impact of adenosine within the TME. Second, we provide an overview of relevant preclinical and clinical data on molecules targeting this pathway, particularly focusing on NSCLC.
Collapse
Affiliation(s)
- Olivier Van Kerkhove
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
| | - Saartje Verfaillie
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Department of Laboratory Medicine, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
- Faculty of Medicine and Life Sciences-LCRC, Hasselt University, 3590 Diepenbeek, Belgium
| | - Kristof Cuppens
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
- Faculty of Medicine and Life Sciences-LCRC, Hasselt University, 3590 Diepenbeek, Belgium
| |
Collapse
|
2
|
Duran I, Banerjee A, Flaherty PJ, Que YA, Ryan CM, Rahme LG, Tsurumi A. Development of a biomarker prediction model for post-trauma multiple organ failure/dysfunction syndrome based on the blood transcriptome. Ann Intensive Care 2024; 14:134. [PMID: 39198331 PMCID: PMC11358370 DOI: 10.1186/s13613-024-01364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Multiple organ failure/dysfunction syndrome (MOF/MODS) is a major cause of mortality and morbidity among severe trauma patients. Current clinical practices entail monitoring physiological measurements and applying clinical score systems to diagnose its onset. Instead, we aimed to develop an early prediction model for MOF outcome evaluated soon after traumatic injury by performing machine learning analysis of genome-wide transcriptome data from blood samples drawn within 24 h of traumatic injury. We then compared its performance to baseline injury severity scores and detection of infections. METHODS Buffy coat transcriptome and linked clinical datasets from blunt trauma patients from the Inflammation and the Host Response to Injury Study ("Glue Grant") multi-center cohort were used. According to the inclusion/exclusion criteria, 141 adult (age ≥ 16 years old) blunt trauma patients (excluding penetrating) with early buffy coat (≤ 24 h since trauma injury) samples were analyzed, with 58 MOF-cases and 83 non-cases. We applied the Least Absolute Shrinkage and Selection Operator (LASSO) and eXtreme Gradient Boosting (XGBoost) algorithms to select features and develop models for MOF early outcome prediction. RESULTS The LASSO model included 18 transcripts (AUROC [95% CI]: 0.938 [0.890-0.987] (training) and 0.833 [0.699-0.967] (test)), and the XGBoost model included 41 transcripts (0.999 [0.997-1.000] (training) and 0.907 [0.816-0.998] (test)). There were 16 overlapping transcripts comparing the two panels (0.935 [0.884-0.985] (training) and 0.836 [0.703-0.968] (test)). The biomarker models notably outperformed models based on injury severity scores and sex, which we found to be significantly associated with MOF (APACHEII + sex-0.649 [0.537-0.762] (training) and 0.493 [0.301-0.685] (test); ISS + sex-0.630 [0.516-0.744] (training) and 0.482 [0.293-0.670] (test); NISS + sex-0.651 [0.540-0.763] (training) and 0.525 [0.335-0.714] (test)). CONCLUSIONS The accurate assessment of MOF from blood samples immediately after trauma is expected to aid in improving clinical decision-making and may contribute to reduced morbidity, mortality and healthcare costs. Moreover, understanding the molecular mechanisms involving the transcripts identified as important for MOF prediction may eventually aid in developing novel interventions.
Collapse
Affiliation(s)
- Ivan Duran
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
| | - Ankita Banerjee
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
| | - Patrick J Flaherty
- Department of Mathematics and Statistics, University of Massachusetts at Amherst, Amherst, MA, 01003, USA
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Colleen M Ryan
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA
- Department of Microbiology and Immunology, Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA, 02115, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA.
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Zhang M, Yang J, Yuan Y, Zhou Y, Wang Y, Cui R, Maliu Y, Xu F, Wu X. Recruitment or activation of mast cells in the liver aggravates the accumulation of fibrosis in carbon tetrachloride-induced liver injury. Mol Immunol 2024; 170:60-75. [PMID: 38626622 DOI: 10.1016/j.molimm.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 04/18/2024]
Abstract
Liver diseases caused by viral infections, alcoholism, drugs, or chemical poisons are a significant health problem: Liver diseases are a leading contributor to mortality, with approximately 2 million deaths per year worldwide. Liver fibrosis, as a common liver disease characterized by excessive collagen deposition, is associated with high morbidity and mortality, and there is no effective treatment. Numerous studies have shown that the accumulation of mast cells (MCs) in the liver is closely associated with liver injury caused by a variety of factors. This study investigated the relationship between MCs and carbon tetrachloride (CCl4)-induced liver fibrosis in rats and the effects of the MC stabilizers sodium cromoglycate (SGC) and ketotifen (KET) on CCl4-induced liver fibrosis. The results showed that MCs were recruited or activated during CCl4-induced liver fibrosis. Coadministration of SCG or KET alleviated the liver fibrosis by decreasing SCF/c-kit expression, inhibiting the TGF-β1/Smad2/3 pathway, depressing the HIF-1a/VEGF pathway, activating Nrf2/HO-1 pathway, and increasing the hepatic levels of GSH, GSH-Px, and GR, thereby reducing hepatic oxidative stress. Collectively, recruitment or activation of MCs is linked to liver fibrosis and the stabilization of MCs may provide a new approach to the prevention of liver fibrosis.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Jinru Yang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yufan Yuan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yan Zhou
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yazhi Wang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Ruirui Cui
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yimai Maliu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Fen Xu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Xin'an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China.
| |
Collapse
|
4
|
Wang P, Liu B, Song C, Jia J, Wang Y, Pang K, Wang Y, Chen C. Exosome MiR-21-5p Upregulated by HIF-1α Induces Adipose Stem Cell Differentiation to Promote Ectopic Bone Formation. Chem Biodivers 2024; 21:e202301972. [PMID: 38342761 DOI: 10.1002/cbdv.202301972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Heterotopic bone occurs after burns, trauma and major orthopedic surgery, which cannot be completely cured by current treatments. The development of new treatments requires more in-depth research into the mechanism of HO. Available evidence suggests that miR-21-5p plays an important role in bone formation. However, its mechanism in traumatic HO is still unclear. First, we identified exosomes extracted from L6 cells using TEM observation of the structure and western blotting detection of the surface marker CD63. Regulation effect of HIF-1α to miR-21-5p was confirmed by q-PCR assay. Then we co-cultured L6 cells with ASCs and performed alizarin red staining and ALP detection. Overexpression of miR-21-5p upregulated BMP4, p-smad1/5/8, OCN and OPN, which suggests the BMP4-smad signaling pathway may be involved in miR-21-5p regulation of osteogenic differentiation of ASCs. Finally in vivo experiments showed that miR-21-5p exosomes promoted ectopic formation in traumatized mice. This study confirms that HIF-1α could modulate miR-21-5p exosomes to promote post-traumatic ectopic bone formation by inducing ASCs cell differentiation. Our study reveals the mechanisms of miR-21-5p in ectopic ossification formation after trauma.
Collapse
Affiliation(s)
- Peng Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Bo Liu
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Chunhao Song
- Department of Medical Imaging, Weihai Wendeng District People Hospital, Weihai, 264200, China
| | - Jun Jia
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Yuanhao Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Kai Pang
- Department of Operations Management, Wehai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Yitao Wang
- Department of Laboratory, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Cong Chen
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| |
Collapse
|
5
|
Signals for Muscular Protein Turnover and Insulin Resistance in Critically Ill Patients: A Narrative Review. Nutrients 2023; 15:nu15051071. [PMID: 36904071 PMCID: PMC10005516 DOI: 10.3390/nu15051071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Sarcopenia in critically ill patients is a highly prevalent comorbidity. It is associated with a higher mortality rate, length of mechanical ventilation, and probability of being sent to a nursing home after the Intensive Care Unit (ICU). Despite the number of calories and proteins delivered, there is a complex network of signals of hormones and cytokines that affect muscle metabolism and its protein synthesis and breakdown in critically ill and chronic patients. To date, it is known that a higher number of proteins decreases mortality, but the exact amount needs to be clarified. This complex network of signals affects protein synthesis and breakdown. Some hormones regulate metabolism, such as insulin, insulin growth factor glucocorticoids, and growth hormone, whose secretion is affected by feeding states and inflammation. In addition, cytokines are involved, such as TNF-alpha and HIF-1. These hormones and cytokines have common pathways that activate muscle breakdown effectors, such as the ubiquitin-proteasome system, calpain, and caspase-3. These effectors are responsible for protein breakdown in muscles. Many trials have been conducted with hormones with different results but not with nutritional outcomes. This review examines the effect of hormones and cytokines on muscles. Knowing all the signals and pathways that affect protein synthesis and breakdown can be considered for future therapeutics.
Collapse
|
6
|
Qin Q, Liu Y, Yang Z, Aimaijiang M, Ma R, Yang Y, Zhang Y, Zhou Y. Hypoxia-Inducible Factors Signaling in Osteogenesis and Skeletal Repair. Int J Mol Sci 2022; 23:ijms231911201. [PMID: 36232501 PMCID: PMC9569554 DOI: 10.3390/ijms231911201] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sufficient oxygen is required to maintain normal cellular and physiological function, such as a creature’s development, breeding, and homeostasis. Lately, some researchers have reported that both pathological hypoxia and environmental hypoxia might affect bone health. Adaptation to hypoxia is a pivotal cellular event in normal cell development and differentiation and in pathological settings such as ischemia. As central mediators of homeostasis, hypoxia-inducible transcription factors (HIFs) can allow cells to survive in a low-oxygen environment and are essential for the regulation of osteogenesis and skeletal repair. From this perspective, we summarized the role of HIF-1 and HIF-2 in signaling pathways implicated in bone development and skeletal repair and outlined the molecular mechanism of regulation of downstream growth factors and protein molecules such as VEGF, EPO, and so on. All of these present an opportunity for developing therapies for bone regeneration.
Collapse
|
7
|
Yu JJ, Non AL, Heinrich EC, Gu W, Alcock J, Moya EA, Lawrence ES, Tift MS, O'Brien KA, Storz JF, Signore AV, Khudyakov JI, Milsom WK, Wilson SM, Beall CM, Villafuerte FC, Stobdan T, Julian CG, Moore LG, Fuster MM, Stokes JA, Milner R, West JB, Zhang J, Shyy JY, Childebayeva A, Vázquez-Medina JP, Pham LV, Mesarwi OA, Hall JE, Cheviron ZA, Sieker J, Blood AB, Yuan JX, Scott GR, Rana BK, Ponganis PJ, Malhotra A, Powell FL, Simonson TS. Time Domains of Hypoxia Responses and -Omics Insights. Front Physiol 2022; 13:885295. [PMID: 36035495 PMCID: PMC9400701 DOI: 10.3389/fphys.2022.885295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The ability to respond rapidly to changes in oxygen tension is critical for many forms of life. Challenges to oxygen homeostasis, specifically in the contexts of evolutionary biology and biomedicine, provide important insights into mechanisms of hypoxia adaptation and tolerance. Here we synthesize findings across varying time domains of hypoxia in terms of oxygen delivery, ranging from early animal to modern human evolution and examine the potential impacts of environmental and clinical challenges through emerging multi-omics approaches. We discuss how diverse animal species have adapted to hypoxic environments, how humans vary in their responses to hypoxia (i.e., in the context of high-altitude exposure, cardiopulmonary disease, and sleep apnea), and how findings from each of these fields inform the other and lead to promising new directions in basic and clinical hypoxia research.
Collapse
Affiliation(s)
- James J. Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Amy L. Non
- Department of Anthropology, Division of Social Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Erica C. Heinrich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Herbert Wertheim School of Public Health and Longevity Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joe Alcock
- Department of Emergency Medicine, University of New Mexico, Albuquerque, MX, United States
| | - Esteban A. Moya
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elijah S. Lawrence
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michael S. Tift
- Department of Biology and Marine Biology, College of Arts and Sciences, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Katie A. O'Brien
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Physiology, Development and Neuroscience, Faculty of Biology, School of Biological Sciences, University of Cambridge, Cambridge, ENG, United Kingdom
| | - Jay F. Storz
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Anthony V. Signore
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Jane I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | | | - Sean M. Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda, CA, United States
| | | | | | | | - Colleen G. Julian
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lorna G. Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Aurora, CO, United States
| | - Mark M. Fuster
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jennifer A. Stokes
- Department of Kinesiology, Southwestern University, Georgetown, TX, United States
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA, United States
| | - John B. West
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jiao Zhang
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - John Y. Shyy
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - Ainash Childebayeva
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - José Pablo Vázquez-Medina
- Department of Integrative Biology, College of Letters and Science, University of California, Berkeley, Berkeley, CA, United States
| | - Luu V. Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - James E. Hall
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Zachary A. Cheviron
- Division of Biological Sciences, College of Humanities and Sciences, University of Montana, Missoula, MT, United States
| | - Jeremy Sieker
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Arlin B. Blood
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jason X. Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Graham R. Scott
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brinda K. Rana
- Moores Cancer Center, UC San Diego, La Jolla, CA, United States
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - Paul J. Ponganis
- Center for Marine Biotechnology and Biomedicine, La Jolla, CA, United States
| | - Atul Malhotra
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Frank L. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
8
|
Fan X, Ren T, Yang W, Zhang X, Yuan L. Activatable photoacoustic/fluorescent dual-modal probe for monitoring of drug-induced liver hypoxia in vivo. Chem Commun (Camb) 2021; 57:8644-8647. [PMID: 34369955 DOI: 10.1039/d1cc02999a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Effective monitoring of liver hypoxia status is crucial for the detection and treatment of drug-induced liver injury. Here, a novel photoacoustic and fluorescent dual-modal probe (NO2-CS) was rationally developed and applied to image isoniazid-induced liver hypoxia through detecting the over-expressed nitroreductase.
Collapse
Affiliation(s)
- Xiaopeng Fan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | | | | | | | | |
Collapse
|
9
|
Han N, Pan Z, Liu G, Yang R, Yujing B. Hypoxia: The "Invisible Pusher" of Gut Microbiota. Front Microbiol 2021; 12:690600. [PMID: 34367091 PMCID: PMC8339470 DOI: 10.3389/fmicb.2021.690600] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022] Open
Abstract
Oxygen is important to the human body. Cell survival and operations depend on oxygen. When the body becomes hypoxic, it affects the organs, tissues and cells and can cause irreversible damage. Hypoxia can occur under various conditions, including external environmental hypoxia and internal hypoxia. The gut microbiota plays different roles under hypoxic conditions, and its products and metabolites interact with susceptible tissues. This review was conducted to elucidate the complex relationship between hypoxia and the gut microbiota under different conditions. We describe the changes of intestinal microbiota under different hypoxic conditions: external environment and internal environment. For external environment, altitude was the mayor cause induced hypoxia. With the increase of altitude, hypoxia will become more serious, and meanwhile gut microbiota also changed obviously. Body internal environment also became hypoxia because of some diseases (such as cancer, neonatal necrotizing enterocolitis, even COVID-19). In addition to the disease itself, this hypoxia can also lead to changes of gut microbiota. The relationship between hypoxia and the gut microbiota are discussed under these conditions.
Collapse
Affiliation(s)
- Ni Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bi Yujing
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
10
|
Elcik D, Tuncay A, Sener EF, Taheri S, Tahtasakal R, Mehmetbeyoğlu E, Gunes I, Emirogullari ON. Blood mRNA Expression Profiles of Autophagy, Apoptosis, and Hypoxia Markers on Blood Cardioplegia and Custodiol Cardioplegia Groups. Braz J Cardiovasc Surg 2021; 36:331-337. [PMID: 33438846 PMCID: PMC8357395 DOI: 10.21470/1678-9741-2020-0330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Introduction: Blood cardioplegia (BC) and Custodiol cardioplegia (CC) have been used for a long time in open heart surgery and are highly effective solutions. The most controversial issue among these two is whether there is any difference between them regarding myocardial damage after ischemia surgery. In this study, autophagy, apoptosis, and hypoxia markers were investigated and that way we evaluated the differences between BC and CC patients. Methods: A total of 30 patients were included in this study, using two different cardioplegic solutions. Three different whole blood samples of the patients were taken from a central vein (preoperatively, immediately postoperatively, and one day after surgery). Total ribonucleic acid was extracted from these samples. Quantitative real-time polymerase chain reaction was performed, and changes in gene expression were determined by the 2-∆∆Ct method of relative quantification. Results: In the CC group, Beclin gene expression level was found to be higher and this difference was statistically significant (P=0.0024). Similarly, cysteine-aspartic acid protease (caspase) 9 and hypoxia-inducible factor 1α messenger ribonucleic acid (mRNA) gene expression level increased and were significantly different in the CC group. In the BC group, Beclin and microtubule-associated protein light chain 3 expressions were higher in the samples taken one day after surgery. Caspases 3 and 8 gene expressions were significantly different in the BC group. Conclusion: As a result of the analysis performed between the two cardioplegia groups, it has been shown that CC harms the myocardium more than BC at the level of mRNA expression of related markers.
Collapse
Affiliation(s)
- Deniz Elcik
- Department of Cardiology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Aydın Tuncay
- Department of Cardiovascular Surgery, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Elif Funda Sener
- Department of Medical Biology, Erciyes University Medical Faculty, Kayseri, Turkey.,Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Serpil Taheri
- Department of Medical Biology, Erciyes University Medical Faculty, Kayseri, Turkey.,Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Reyhan Tahtasakal
- Department of Medical Biology, Erciyes University Medical Faculty, Kayseri, Turkey.,Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Ecmel Mehmetbeyoğlu
- Department of Medical Biology, Erciyes University Medical Faculty, Kayseri, Turkey.,Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Isın Gunes
- Department of Anesthesiology and Reanimation, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Omer Naci Emirogullari
- Department of Cardiovascular Surgery, Erciyes University Medical Faculty, Kayseri, Turkey
| |
Collapse
|
11
|
Cui H, Yang A, Zhou H, Wang Y, Luo J, Zhou J, Liu T, Li P, Zhou J, Hu E, He Z, Hu W, Tang T. Thrombin-induced miRNA-24-1-5p upregulation promotes angiogenesis by targeting prolyl hydroxylase domain 1 in intracerebral hemorrhagic rats. J Neurosurg 2021; 134:1515-1526. [PMID: 32413855 DOI: 10.3171/2020.2.jns193069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/24/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Thrombin is a unique factor that triggers post-intracerebral hemorrhage (ICH) angiogenesis by increasing hypoxia-inducible factor-1α (HIF-1α) at the protein level. However, HIF-1α mRNA remains unchanged. MicroRNAs (miRNAs) mediate posttranscriptional regulation by suppressing protein translation from mRNAs. This study aimed to determine if miRNAs might be involved in thrombin-induced angiogenesis after ICH by targeting HIF-1α or its upstream prolyl hydroxylase domains (PHDs). METHODS The study was divided into two parts. In part 1, rats received an injection of thrombin into the right globus pallidus. An miRNA array combined with miRNA target prediction, luciferase activity assay, and miRNA mimic/inhibitor transfection were used to identify candidate miRNAs and target genes. Part 2 included experiments 1 and 2. In experiment 1, rats were randomly divided into the sham group, ICH group, and ICH+hirudin-treated (thrombin inhibitor) group. In experiment 2, the rats were randomly divided into the sham group, ICH group, ICH+antagomir group, ICH+antagomir-control group, and ICH+vehicle group. Western blotting and quantitative real-time polymerase chain reaction were used to determine the expression of protein and miRNA, respectively. The coexpression of miR-24-1-5p (abbreviated to miR-24) and von Willebrand factor was detected by in situ hybridization and immunohistochemical analysis. The angiogenesis was evaluated by double-labeling immunofluorescence. Neurological function was evaluated by body weight, modified Neurological Severity Scores, and corner turn and foot-fault tests. RESULTS In part 1, it was shown that miR-24, which is predicted to target PHD1, was upregulated (fold-change of 1.83) after thrombin infusion, and that the miR-24 mimic transfection decreased luciferase activity and downregulated PHD1 expression (p < 0.05). miR-24 inhibitor transfection increased PHD1 expression (p < 0.05). In part 2, it was shown that miR-24 was expressed in endothelial cells. The HIF-1α protein level and proliferating cell nuclear antigen-positive (PCNA+) nuclei in vessels were increased, while the PHD1 protein level was decreased after ICH, and these effects were reversed by hirudin (p < 0.05). The antagomiR-24-treated rats exhibited a markedly lower body weight and significantly poorer recovery from neurological deficit compared with those in ICH groups (p < 0.05). AntagomiR-24 intervention also led to lower miR-24 expression, a higher PHD1 protein level, and fewer PCNA+ nuclei in vessels compared with those in ICH groups (p < 0.05). CONCLUSIONS The present study suggests that thrombin reduces HIF-1α degradation and initiates angiogenesis by increasing miR-24, which targets PHD1 after ICH.
Collapse
Affiliation(s)
| | - Ali Yang
- 2Department of Neurology, Henan Province People's Hospital, Zhengzhou; and
| | - Huajun Zhou
- 3Institute of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Yang Wang
- 1Institute of Integrative Medicine and
| | | | - Jun Zhou
- 4Institute of Medical Science, Xiangya Hospital, Central South University, Changsha, Hunan
| | - Tao Liu
- 1Institute of Integrative Medicine and
| | | | - Jing Zhou
- 1Institute of Integrative Medicine and
| | - En Hu
- 1Institute of Integrative Medicine and
| | - Zehui He
- 1Institute of Integrative Medicine and
| | - Wang Hu
- 1Institute of Integrative Medicine and
| | - Tao Tang
- 1Institute of Integrative Medicine and
| |
Collapse
|
12
|
Woods P, Alcock J. High-altitude pulmonary edema. Evol Med Public Health 2021; 9:118-119. [PMID: 33732460 PMCID: PMC7947961 DOI: 10.1093/emph/eoaa052] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Lay summary: High Altitude Pulmonary Edema (HAPE) is a potentially fatal disease of altitude, in which the lungs can become filled with fluid. In this article we explore the mechanisms causing this condition and how it can be viewed as a condition of a mismatch between current environment and evolutionary experience.
Collapse
Affiliation(s)
- Peter Woods
- Department of Respiratory Medicine, University Hospitals Coventry and Warwickshire, Coventry, UK
| | - Joe Alcock
- Department of Emergency Medicine, University of New Mexico, Albuquerque, USA
| |
Collapse
|
13
|
Li LF, Yu CC, Huang HY, Wu HP, Chu CM, Huang CY, Liu PC, Liu YY. Suppression of Hypoxia-Inducible Factor 1α by Low-Molecular-Weight Heparin Mitigates Ventilation-Induced Diaphragm Dysfunction in a Murine Endotoxemia Model. Int J Mol Sci 2021; 22:ijms22041702. [PMID: 33567713 PMCID: PMC7914863 DOI: 10.3390/ijms22041702] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanical ventilation (MV) is required to maintain life for patients with sepsis-related acute lung injury but can cause diaphragmatic myotrauma with muscle damage and weakness, known as ventilator-induced diaphragm dysfunction (VIDD). Hypoxia-inducible factor 1α (HIF-1α) plays a crucial role in inducing inflammation and apoptosis. Low-molecular-weight heparin (LMWH) was proven to have anti-inflammatory properties. However, HIF-1α and LMWH affect sepsis-related diaphragm injury has not been investigated. We hypothesized that LMWH would reduce endotoxin-augmented VIDD through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α–deficient, were exposed to MV with or without endotoxemia for 8 h. Enoxaparin (4 mg/kg) was administered subcutaneously 30 min before MV. MV with endotoxemia aggravated VIDD, as demonstrated by increased interleukin-6 and macrophage inflammatory protein-2 levels, oxidative loads, and the expression of HIF-1α, calpain, caspase-3, atrogin-1, muscle ring finger-1, and microtubule-associated protein light chain 3-II. Disorganized myofibrils, disrupted mitochondria, increased numbers of autophagic and apoptotic mediators, substantial apoptosis of diaphragm muscle fibers, and decreased diaphragm function were also observed (p < 0.05). Endotoxin-exacerbated VIDD and myonuclear apoptosis were attenuated by pharmacologic inhibition by LMWH and in HIF-1α–deficient mice (p < 0.05). Our data indicate that enoxaparin reduces endotoxin-augmented MV-induced diaphragmatic injury, partially through HIF-1α pathway inhibition.
Collapse
Affiliation(s)
- Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Hung-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Huang-Pin Wu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chien-Ming Chu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Chi Liu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Correspondence: ; Tel.: +(886)-2-28712121 (ext. 3071); Fax: +(886)-2-28757858
| |
Collapse
|
14
|
Li X, Berg NK, Mills T, Zhang K, Eltzschig HK, Yuan X. Adenosine at the Interphase of Hypoxia and Inflammation in Lung Injury. Front Immunol 2021; 11:604944. [PMID: 33519814 PMCID: PMC7840604 DOI: 10.3389/fimmu.2020.604944] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Hypoxia and inflammation often coincide in pathogenic conditions such as acute respiratory distress syndrome (ARDS) and chronic lung diseases, which are significant contributors to morbidity and mortality for the general population. For example, the recent global outbreak of Coronavirus disease 2019 (COVID-19) has placed viral infection-induced ARDS under the spotlight. Moreover, chronic lung disease ranks the third leading cause of death in the United States. Hypoxia signaling plays a diverse role in both acute and chronic lung inflammation, which could partially be explained by the divergent function of downstream target pathways such as adenosine signaling. Particularly, hypoxia signaling activates adenosine signaling to inhibit the inflammatory response in ARDS, while in chronic lung diseases, it promotes inflammation and tissue injury. In this review, we discuss the role of adenosine at the interphase of hypoxia and inflammation in ARDS and chronic lung diseases, as well as the current strategy for therapeutic targeting of the adenosine signaling pathway.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Tianjin Medical University NanKai Hospital, Tianjin, China
| | - Nathanial K. Berg
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Mills
- Department of Biochemistry, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kaiying Zhang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
15
|
Abstract
The syndrome of critical illness is a complex physiological stressor that can be triggered by diverse pathologies. It is widely believed that organ dysfunction and death result from bioenergetic failure caused by inadequate cellular oxygen supply. Teleologically, life has evolved to survive in the face of stressors by undergoing a suite of adaptive changes. Adaptation not only comprises alterations in systemic physiology but also involves molecular reprogramming within cells. The concept of cellular adaptation in critically ill patients is a matter of contention in part because medical interventions mask underlying physiology, creating the artificial construct of "chronic critical illness," without which death would be imminent. Thus far, the intensive care armamentarium has not targeted cellular metabolism to preserve a temporary equilibrium but instead attempts to normalize global oxygen and substrate delivery. Here, we review adaptations to hypoxia that have been demonstrated in cellular models and in human conditions associated with hypoxia, including the hypobaric hypoxia of high altitude, the intrauterine low-oxygen environment, and adult myocardial hibernation. Common features include upregulation of glycolytic ATP production, enhancement of respiratory efficiency, downregulation of mitochondrial density, and suppression of energy-consuming processes. We argue that these innate cellular adaptations to hypoxia represent potential avenues for intervention that have thus far remained untapped by intensive care medicine.
Collapse
Affiliation(s)
- Helen T McKenna
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Royal Free Intensive Care Unit, Royal Free Hospital, London, United Kingdom
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Daniel S Martin
- Royal Free Intensive Care Unit, Royal Free Hospital, London, United Kingdom.,Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
16
|
Yan Z, Xu G. A Novel Choice to Correct Inflammation-Induced Anemia in CKD: Oral Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat. Front Med (Lausanne) 2020; 7:393. [PMID: 32850902 PMCID: PMC7423837 DOI: 10.3389/fmed.2020.00393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
Anemia is a complication of chronic kidney disease (CKD), primarily due to insufficient secretion of erythropoietin (EPO) by the kidney. Erythropoiesis-stimulating agents (ESAs) are used to treat anemia associated with chronic kidney disease. A poor response to ESAs has been associated with inflammation. Inflammation can affect erythrocytes and its production in many ways, but mainly through the inflammatory cytokine IL-6 to stimulate the synthesis of hepcidin in the liver. Hepcidin causes iron insufficiency, which causes erythrocytes to fail to mature normally. In addition, inhibition of bone marrow erythroid precursor cells by inflammatory cytokines such as IL-1 and TNF-α also affects bone marrow hematopoiesis. These cytokines are also important factors leading to EPO resistance. Roxadustat is a new drug for the treatment of renal anemia. In addition to promoting the production of EPO, clinical trials have shown that it can significantly reduce hepcidin and can potentially be used for the treatment of inflammation-induced anemia in CKD.
Collapse
Affiliation(s)
- Zhipeng Yan
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Yu Y, Ma L, Zhang H, Sun W, Zheng L, Liu C, Miao L. EPO could be regulated by HIF-1 and promote osteogenesis and accelerate bone repair. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:206-217. [PMID: 31851837 DOI: 10.1080/21691401.2019.1699827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bone defects caused by many factors prompt further study of pathological process and restoration methods. This study was aimed to clarify the effect of erythropoietin on the repair of bone defect. We added the designated concentration of rhEPO to endothelial progenitor cells and marrow stromal cells, then detected its osteogenic and angiogenesis effects. The results showed that rhEPO promoted the proliferation of EPC and ST2 by promoting the mitosis without affecting cell apoptosis. The protein and mRNA levels of angiogenesis and osteogenic related factors exhibited higher expressions. Additionally, rhEPO encapsulated in PLGA scaffolds accelerated the new bone formation in rat calvaria bone defect model. Since the centre of bone defect was hypoxia environment, we cultured EPC and ST2 under hypoxia. SiRNA and an inhibitor of HIF-1 were used to interfere HIF-1, then the following changes of VEGF and EPO were detected. The results showed that all the factors were upregulated under the hypoxia environment. The expression of VEGF at protein and mRNA level decreased as HIF-1 was inhibited or interfered from 6 h, while the mRNA expression of EPO from 6 h and changed significantly at protein level from 12 h. Therefore, EPO is a promising factor for further studies.
Collapse
Affiliation(s)
- Yijun Yu
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Lan Ma
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - He Zhang
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Weibin Sun
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Lichun Zheng
- Department of Preventive Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Chao Liu
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Leiying Miao
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, PR China
| |
Collapse
|
18
|
Low-Molecular-Weight Heparin Reduces Ventilation-Induced Lung Injury through Hypoxia Inducible Factor-1α in a Murine Endotoxemia Model. Int J Mol Sci 2020; 21:ijms21093097. [PMID: 32353952 PMCID: PMC7247708 DOI: 10.3390/ijms21093097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.
Collapse
|
19
|
Huang Y, Wang X, Lin H. The hypoxic microenvironment: a driving force for heterotopic ossification progression. Cell Commun Signal 2020; 18:20. [PMID: 32028956 PMCID: PMC7006203 DOI: 10.1186/s12964-020-0509-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/02/2020] [Indexed: 12/23/2022] Open
Abstract
Heterotopic ossification (HO) refers to the formation of bone tissue outside the normal skeletal system. According to its pathogenesis, HO is divided into hereditary HO and acquired HO. There currently lack effective approaches for HO prevention or treatment. A deep understanding of its pathogenesis will provide promising strategies to prevent and treat HO. Studies have shown that the hypoxia-adaptive microenvironment generated after trauma is a potent stimulus of HO. The hypoxic microenvironment enhances the stability of hypoxia-inducible factor-1α (HIF-1α), which regulates a complex network including bone morphogenetic proteins (BMPs), vascular endothelial growth factor (VEGF), and neuropilin-1 (NRP-1), which are implicated in the formation of ectopic bone. In this review, we summarize the current understanding of the triggering role and underlying molecular mechanisms of the hypoxic microenvironment in the initiation and progression of HO, focusing mainly on HIF-1 and it's influenced genes BMP, VEGF, and NRP-1. A better understanding of the role of hypoxia in HO unveils novel therapeutic targets for HO that reduce the local hypoxic microenvironment and inhibit HIF-1α activity. Video Abstract. (MP4 52403 kb)
Collapse
Affiliation(s)
- Yifei Huang
- First Clinical Medical School, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xinyi Wang
- First Clinical Medical School, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 461 BaYi Avenue, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
20
|
Comparative Evaluation of the Angiogenic Potential of Hypoxia Preconditioned Blood-Derived Secretomes and Platelet-Rich Plasma: An In Vitro Analysis. Biomedicines 2020; 8:biomedicines8010016. [PMID: 31963131 PMCID: PMC7168246 DOI: 10.3390/biomedicines8010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Blood-derived factor preparations are being clinically employed as tools for promoting tissue repair and regeneration. Here we set out to characterize the in vitro angiogenic potential of two types of frequently used autologous blood-derived secretomes: platelet-rich plasma (PRP) and hypoxia preconditioned plasma (HPP)/serum (HPS). The concentration of key pro-angiogenic (VEGF) and anti-angiogenic (TSP-1, PF-4) protein factors in these secretomes was analyzed via ELISA, while their ability to induce microvessel formation and sprouting was examined in endothelial cell and aortic ring cultures, respectively. We found higher concentrations of VEGF in PRP and HPP/HPS compared to normal plasma and serum. This correlated with improved induction of microvessel formation by PRP and HPP/HPS. HPP had a significantly lower TSP-1 and PF-4 concentration than PRP and HPS. PRP and HPP/HPS appeared to induce similar levels of microvessel sprouting; however, the length of these sprouts was greater in HPP/HPS than in PRP cultures. A bell-shaped angiogenic response profile was observed with increasing HPP/HPS dilutions, with peak values significantly exceeding the PRP response. Our findings demonstrate that optimization of peripheral blood cell-derived angiogenic factor signalling through hypoxic preconditioning offers an improved alternative to simple platelet concentration and release of growth factors pre-stored in platelets.
Collapse
|
21
|
Identifying Synergistic Mechanisms of Multiple Ingredients in Shuangbai Tablets against Proteinuria by Virtual Screening and a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1027271. [PMID: 32025234 PMCID: PMC6984745 DOI: 10.1155/2020/1027271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/08/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
Shuangbai Tablets (SBT), a traditional herbal mixture, has shown substantial clinical efficacy. However, a systematic mechanism of its active ingredients and pharmacological mechanisms of action against proteinuria continues being lacking. A network pharmacology approach was effectual in discovering the relationship of multiple ingredients and targets of the herbal mixture. This study aimed to identify key targets, major active ingredients, and pathways of SBT against proteinuria by network pharmacology approach combined with thin layer chromatography (TLC). Human phenotype (HP) disease analysis, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and molecular docking were used in this study. To this end, a total of 48 candidate targets of 118 active ingredients of SBT were identified. Network analysis showed PTGS2, ESR1, and NOS2 to be the three key targets, and beta-sitosterol, quercetin, and berberine were the three major active ingredients; among them one of the major active ingredients, quercetin, was discriminated by TLC. These results of the functional enrichment analysis indicated that the most relevant disease including these 48 candidate proteins is proteinuria, SBT treated proteinuria by sympathetically regulating multiple biological pathways, such as the HIF-1, RAS, AGE-RAGE, and VEGF signaling pathways. Additionally, molecular docking validation suggested that major active ingredients of SBT were capable of binding to HIF-1A and VEGFA of the main pathways. Consequently, key targets, major active ingredients, and pathways based on data analysis of SBT against proteinuria were systematically identified confirming its utility and providing a new drug against proteinuria.
Collapse
|
22
|
Relationship between HIF-1α and apoptosis in rats with traumatic brain injury and the influence of traditional Chinese medicine Sanqi. Saudi J Biol Sci 2020; 26:1995-1999. [PMID: 31889784 PMCID: PMC6923447 DOI: 10.1016/j.sjbs.2019.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/02/2019] [Accepted: 08/04/2019] [Indexed: 11/20/2022] Open
Abstract
Objective To explore the expression of HIF-1α, neuronal apoptosis and the influence of traditional Chinese medicine Sanqi on hematoma after brain injury in rats. Methods Ninety SD rats were divided into 3 groups randomly: blank control group, traumatic brain injury (TBI) group and Sanqi intervention group, and they were decapitated after brain injury at different time points: 6 h, 1 d, 2 d, 3 d, 5 d, 7 d. The model of cerebral hemorrhage was made by autologous non-coagulation in stereotactic locator, the expression of HIF-1α and TUNEL-positive cells (apoptotic cells) in the perihematomal area was detected by immunohistochemistry. Results In blank control group, a small amount of HIF-1α was expressed and apoptotic cells were observed. The expression of HIF-1α was up-regulated in the brain injury group from 6 h, and the apoptotic cells increased in abundance. The peak of HIF-1α was reached at 3 d, then decreased, and remained at the high level on the 7 d. Compared with blank control group, the TBI group was statistically significant (P < 0.05). The Chinese medicine Sanqi intervention group significantly up-regulated HIF-1α'expression and decreased neuronal apoptosis, which was statistically significant (P < 0.05). Conclusion HIF-1α's expression was up-regulated around the hematoma after brain injury, and the apoptosis of nerve cells was obviously increased. The traditional Chinese medicine Sanqi can significantly increase the expression of HIF-1α, reduce the apoptosis around the hematoma, and thus play a neuroprotective role.
Collapse
|
23
|
Meng C, Sun Y, Hu Z, Wang H, Jiang W, Song J, Yu Y, Hu D. Effects of hypoxia inducible factor-1α on expression levels of MLCK, p-MLC and ZO-1 of rat endothelial cells. Biochem Biophys Res Commun 2019; 519:591-596. [PMID: 31540688 DOI: 10.1016/j.bbrc.2019.08.159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/30/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To examine the aberrant expression of endothelial permeability associated proteins including MLCK, p-MLC and ZO-1 in presence of different levels of hypoxia-inducible factor 1 alpha (HIF-1α). METHODS We established monolayer vascular endothelial cell model with the primary rat endothelial cells. Over-expressed or under-expressed HIF-1α cell lines were made by endothelial cells transfected with plasmid vector constructed with HIF-1α gene or HIF-1α-specific short hairpin RNA (shRNA). Levels of mRNA and protein of MLCK, p-MLC and ZO-1 were determined using Real-Time PCR and Western blot. All data were analyzed using by One-Way ANOVA method and LSD. RESULTS We successfully cultured the rat endothelial primary cells for four days. The mRNA and protein levels of MLCK and p-MLC were significantly increased in the HIF-1α over-expression group than that in the blank control group and the empty plasmid GV230 group (P<0.05). ZO-1 was significantly lower in the HIF-1α over-expression group than that in the blank control group and the GV230 group. On the contrary, the mRNA and protein levels of MLCK and p-MLC were significantly lower in the HIF-1α under-expression group than that in the blank control group and the shRNA-NC group (P<0.05). ZO-1 was significantly higher in the HIF-1α low-expression group than that in the blank control group and the shRNA-NC group. CONCLUSION HIF-1α positively regulates the expression of MLCK and p-MLC and negatively regulates the expression of ZO-1 in rat monolayer endothelial cells.
Collapse
Affiliation(s)
- Chengying Meng
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Youjun Sun
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zijian Hu
- 2018 Class of Clinical Medicine (No.1813010207), The First Clinical College of Anhui Medical University, Hefei, 230022, China
| | - Huan Wang
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Jiang
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Junhui Song
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Youxin Yu
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Delin Hu
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|