1
|
Zhang W, Jiang L, Tong X, He H, Zheng Y, Xia Z. Sepsis-Induced Endothelial Dysfunction: Permeability and Regulated Cell Death. J Inflamm Res 2024; 17:9953-9973. [PMID: 39628705 PMCID: PMC11612565 DOI: 10.2147/jir.s479926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Endothelial cells (ECs) are an important cell type typically affected in sepsis, resulting in compromised barrier function and various forms of regulated cell death (RCD). However, the precise mechanisms underlying sepsis-induced EC damage remain unclear. This review summarizes the recent research progress on factors and mechanisms that may affect the permeability and RCD of ECs under septic conditions, including glycocalyx, damage-associated molecular patterns, and various forms of RCD in ECs, such as apoptosis, pyroptosis, ferroptosis, and autophagy. This review offers important insights into the underlying mechanisms of endothelial dysfunction in sepsis, aiming to contribute to developing small-molecule targeted clinical therapies.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Xirui Tong
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Heng He
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
- Research Unit of Key Techniques for Treatment of burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
2
|
Yang Y, Li N, Song J, Tian Y, Chen B, Li J, Lin L, Qin Z. Hemolysis-associated release of hemoglobin induces mitochondrial oxidative phosphorylation (OXPHOS) disturbance and aggravates cell oxidative damage in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2024; 157:110043. [PMID: 39592030 DOI: 10.1016/j.fsi.2024.110043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024]
Abstract
The liver is a key site for the removal of cell-free hemin during hemolysis. However, the mechanism underlying liver damage caused by hemolysis in teleost hemolytic disorderss remains unclear. In this study, the hemin incubation of grass carp liver cells (L8824) and phenylhydrazine (PHZ) injection were employed to simulate in vitro and in vivo hemolysis models. The Cell Counting Kit (CCK) assay results of the L8824 cells showed that the hemin caused obvious cell death and exhibited concentration-dependent characteristics. Furthermore, hemin stimulation significantly increased intracellular iron content, markedly enhanced intracellular ROS (reactive oxygen species) production, triggered the activation of genes linked to iron metabolism, and disrupted mitochondrial structural integrity. The quantitative real-time PCR (qRT-PCR) assay and enzyme activity findings indicated that the hemoglobin (Hb) treatment activated the activity and expression of mitochondrial respiratory chain complexes, while the addition of compound inhibitors I, II, and III could rescue hemin-induced cell death. Finally, a hemolysis model was established via intraperitoneal injection of PHZ in the grass carp. Histopathological analysis and in vivo transcriptome data showed that PHZ-induced hemolysis resulted in liver inflammation and iron and collagen fiber buildup. Additionally, immunofluorescence and immunohistochemical data indicated it enhanced the ROS generation, malondialdehyde (MDA), and 4-hydroxy-2-nonenal (4-HNE), destroyed the mitochondria, and up-regulated the transcription of mitochondrial respiratory chain complexes. In summary, the cell-free Hb released during hemolysis increased iron deposition, disrupted iron metabolism homeostasis, and caused oxidative stress. Consequently, this destroyed mitochondria function and ultimately exacerbated cell death.
Collapse
Affiliation(s)
- Yan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ningjing Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Jialing Song
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ye Tian
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Bing Chen
- Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Jiangtao Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
3
|
García-Caballero C, Guerrero-Hue M, Vallejo-Mudarra M, Palomino Antolin A, Decouty-Pérez C, Sánchez-Mendoza LM, Villalba JM, González-Reyes JA, Opazo-Rios L, Vázquez-Carballo C, Herencia C, Leiva-Cepas F, Cortegano I, Andrés BD, Egido J, Egea J, Moreno JA. Nox4 is involved in acute kidney injury associated to intravascular hemolysis. Free Radic Biol Med 2024; 225:430-444. [PMID: 39413979 DOI: 10.1016/j.freeradbiomed.2024.10.283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Massive intravascular hemolysis occurs not unfrequently in many clinical conditions. Breakdown of erythrocytes promotes the accumulation of heme-derivates in the kidney, increasing oxidative stress and cell death, thus promoting acute kidney injury (AKI). NADPH oxidase 4 (Nox4) is a major source of reactive oxygen species (ROS) in the kidney, however it is unknown the role of Nox4 in hemolysis and whether inhibition of this enzyme may protect from heme-mediated injury. To answer these questions, we elicited intravascular hemolysis in wild type and Nox4 knockout mice. We also evaluated whether nephrotoxic effects of heme may be reduced by using Nox4 siRNA and pharmacologic inhibition with GKT137831, a Nox4 inhibitor, both in vivo and in cultured renal cells. Our results showed that induction of massive hemolysis elicited AKI characterized by loss of renal function, morphological alterations of the tubular epithelium and podocytes, oxidative stress, inflammation, mitochondrial dysfunction, blockade of autophagy and cell death. These pathological effects were significantly prevented in Nox4-deficient mice and in animals treated with GKT137831. In vitro studies showed that Nox4 disruption by specific siRNAs or Nox4 inhibitors declined heme-mediated ROS production and cell death. Our data identify Nox4 as a key enzyme involved in intravascular hemolysis-induced AKI. Thus, Nox4 inhibition may be a potential therapeutic approach to prevent renal damage in patients with severe hemolytic crisis.
Collapse
Affiliation(s)
- Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Mercedes Vallejo-Mudarra
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Alejandra Palomino Antolin
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Celine Decouty-Pérez
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Luz Marina Sánchez-Mendoza
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - José Manuel Villalba
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - José Antonio González-Reyes
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - Lucas Opazo-Rios
- Health Science Faculty, University of Las Américas, Concepción, Talcahuano, Chile.
| | - Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain.
| | - Carmen Herencia
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain.
| | - Fernando Leiva-Cepas
- Departament of Morphological and Sociosanitary Sciences, Pathology Unit, Faculty of Medicine and Nurse, University of Cordoba/Pathology Unit, Hospital Universitario Reina Sofía, Cordoba, Spain.
| | - Isabel Cortegano
- Immunobiology Department, Carlos III Health Institute, Madrid, Spain.
| | - Belén de Andrés
- Immunobiology Department, Carlos III Health Institute, Madrid, Spain.
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| | - Javier Egea
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| |
Collapse
|
4
|
Wang ZX, Jiao WJ, Yang Y, Liu HL, Wang HL. Role of inflammasomes in Toxoplasma and Plasmodium infections. Parasit Vectors 2024; 17:466. [PMID: 39548522 PMCID: PMC11566176 DOI: 10.1186/s13071-024-06529-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/08/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The detection of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by multimeric protein complexes, known as inflammasomes, triggers an inflammatory response, which is a critical component of the innate immune system. This inflammatory response plays a pivotal role in host resistance against parasitic infections, presenting a significant global health challenge. METHODS We systematically searched for relevant articles from the Pubmed and the Web of Science database to summarize current insights into how inflammasomes function in preventing infections caused by the apicomplexan parasites Toxoplasma and Plasmodium. RESULTS In vivo and in vitro studies have extensively explored inflammasomes such as the absent in melanoma 2 (AIM2), NLR family pyrin-containing protein 1 (NLRP1), NLRP3, and NLRP12 inflammasomes, alongside noncanonical inflammasomes, with particular emphasis on the NLRP1 and the NLRP3 inflammasome during Toxoplasma gondii infection or the AIM2 and the NLRP3 inflammasome at various stages of Plasmodium infection. Toxoplasma gondii interacts with inflammasomes to activate or inhibit immune responses. CONCLUSIONS Inflammasomes control parasite burden and parasite-induced cell death, contribute to immune recognition and inflammatory responses and thus influence apicomplexan parasite-associated pathogenesis and the severity of clinical outcomes. Hence, inflammasomes play crucial roles in the progression and outcomes of toxoplasmosis and malaria. A comprehensive understanding of how parasitic infections modulate inflammasome activity enhances insight into host immune responses against parasites.
Collapse
Affiliation(s)
- Zhi-Xin Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Wan-Jun Jiao
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Yong Yang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Hong-Li Liu
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China.
| | - Hai-Long Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China.
| |
Collapse
|
5
|
Navaneethabalakrishnan S, An X, Vinchi F. Heme- and iron-activated macrophages in sickle cell disease: an updated perspective. Curr Opin Hematol 2024; 31:275-284. [PMID: 39046855 PMCID: PMC11427154 DOI: 10.1097/moh.0000000000000836] [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: 07/27/2024]
Abstract
PURPOSE OF REVIEW Sickle cell disease (SCD) is a hereditary blood disorder due to a single-point mutation in the β-globin gene. The ensuing hemoglobin has the tendency to polymerize upon deoxygenation, leading to the typical sickle shape of red blood cells. While the primary pathology of sickle cell disease is a direct consequence of altered red blood cells, emerging evidence highlights the central role of macrophages in mediating hemoglobin scavenging, perpetuating oxidative stress and inflammation, and causing endothelial dysfunction and tissue remodeling. RECENT FINDINGS Recent research uncovered the impact of heme and iron overload on macrophage polarization and functions in sickle cell disease, and its implication for chronic inflammation and tissue damage in vital organs such as the liver, spleen, lungs and kidneys. By providing a thorough understanding of the dynamic interactions between macrophages and various cellular components within the sickle cell disease milieu, these studies have laid the foundation for the identification of macrophage-related cellular and molecular mechanisms potentially targetable for therapeutic purposes to attenuate sickle complications. SUMMARY This review provides a current update about recent discoveries on heme/iron-activated macrophages in SCD, shedding light on their critical role in disease pathophysiology. Ultimately, it proposes avenues for future research aimed at addressing the relevance of these cells for other sickle complications and at targeting them to mitigate disease morbidity and improve patient outcomes.
Collapse
Affiliation(s)
| | - Xiuli An
- Laboratory of Membrane Biology, Lindsley Kimball Research Institute, New York Blood Center
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley Kimball Research Institute, New York Blood Center
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
6
|
Lucas D, Munoz C, O'Boyle Q, Pires IS, Palmer AF, Cabrales P. Mitigating hemoglobin-induced nephropathy: ApoHb-hp protection of podocytes. Physiol Rep 2024; 12:e70132. [PMID: 39578364 PMCID: PMC11584305 DOI: 10.14814/phy2.70132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/11/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
This study investigates hemoglobin (Hb)-induced kidney injury and the protective role of the ApoHemoglobin-Haptoglobin (ApoHb-Hp) complex against heme and Hb damage. Hb facilitates oxygen (O2) delivery but poses challenges outside red blood cells (RBCs) due to toxic Hb and heme mechanisms. These are managed by binding to serum proteins like Haptoglobin (Hp) and Hemopexin (Hpx). During hemolysis, depletion of Hp and Hpx leaves tissues vulnerable to Hb and heme. To address this, we developed the ApoHb-Hp complex, based on Apohemoglobin, which is produced by removing heme from Hb, conjugated with Hp. This complex acts as a dual scavenger for Hb and heme, preventing tissue damage. Our findings demonstrate that ApoHb-Hp significantly protects MPC5 podocytes from Hb-induced damage. Fluorescent staining showed a higher percentage of nephrin-positive cells in the ApoHb-Hp group, and MTT assays revealed enhanced cell viability compared to Hb alone. Additionally, ApoHb-Hp reduced reactive oxygen species (ROS) production, with the Hb group exhibiting significantly elevated ROS levels. The ApoHb-Hp complex mitigated the depletion of protective mechanisms, as shown by significant increases in superoxide dismutase (SOD) and glutathione (GSH). Moreover, ApoHb-Hp treatment reduced the activation of the NLRP3 inflammasome signaling pathway and inflammatory cytokines IL-1β and IL-18. These findings underscore the therapeutic potential of ApoHb-Hp in mitigating Hb-induced renal damage by preserving podocyte viability and reducing oxidative stress. Overall, ApoHb-Hp maintained protective mechanisms depleted otherwise by Hb. These findings highlight ApoHb-Hp's potential as a therapeutic agent against Hb-induced renal damage, offering insights into its mechanisms and implications for treating conditions involving hemolysis.
Collapse
Affiliation(s)
- Daniela Lucas
- Department of BioengineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Carlos Munoz
- Department of BioengineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Quintin O'Boyle
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Ivan S. Pires
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Andre F. Palmer
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Pedro Cabrales
- Department of BioengineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| |
Collapse
|
7
|
Gao F, Zeng S, Chao D, Wu L. Dl-3-n-Butylphthalide attenuates early brain injury and delayed neurological dysfunction by regulating NLRP3 inflammasome after subarachnoid hemorrhage. Brain Res Bull 2024; 217:111084. [PMID: 39304001 DOI: 10.1016/j.brainresbull.2024.111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
Subarachnoid hemorrhage (SAH) is a severe neurological event lacking of effective therapy. Early brain injury (EBI) and delayed neurological dysfunction are important cause in the poor prognosis of patients with SAH. Nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome activation has been implicated in many inflammatory lesion pathogeneses including SAH. Dl-3-n-butylphthalide (NBP) has been reported to possess substantial anti-inflammatory properties, which is beneficial for various neurodegenerative diseases. However, the effect and molecular mechanisms of NBP on SAH have not been clearly identified. We designed this study to investigate the effect of NBP against EBI and delayed neurological dysfunction after SAH and to reveal the possible underlying mechanism. The adult mice were subjected to endovascular perforation SAH model or sham operation. Mice were randomized to sham group, SAH group, or SAH+NBP group. The EBI (short-term study) was studied at 48 h post-SAH and delayed neurological dysfunction (long-term study) at 21 days post-SAH. The results suggested that NBP evidently alleviated the EBI in mice at 48 h post-SAH, as shown by elevating neurological score, reducing brain edema, blood-brain barrier disruption, neuronal loss, and astrocyte aggregation, as well as ameliorating cerebral vasospasm. Moreover, NBP was able to improve long-term neurobehavioral functions and decrease neuronal apoptosis at 21 days after SAH. Significantly, NBP treatment also inhibited the expressions of NLRP3, ASC, caspase-1, cleaved-caspase-1, IL-1β, IL-18, GSDMD and GSDMD-N in both EBI and delayed neurological dysfunction induced by SAH. Our findings suggested that NBP treatment exerts a profound neuroprotective effect against early brain injury and delayed neurological dysfunction induced by SAH, at least partially through regulating NLRP3 inflammasome signaling pathway and its related inflammation and pyroptosis.
Collapse
Affiliation(s)
- Fangfang Gao
- Nanshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Shenzhen Nanshan Hospital of Chinese Medicine), Shenzhen 518052, China
| | - Shujin Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dachong Chao
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou 510006, China
| | - Liangmiao Wu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Huang Y, Jiang W, Zhou R. DAMP sensing and sterile inflammation: intracellular, intercellular and inter-organ pathways. Nat Rev Immunol 2024; 24:703-719. [PMID: 38684933 DOI: 10.1038/s41577-024-01027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules that are released from host cells as a result of cell death or damage. The release of DAMPs in tissues is associated with loss of tissue homeostasis. Sensing of DAMPs by innate immune receptors triggers inflammation, which can be beneficial in initiating the processes that restore tissue homeostasis but can also drive inflammatory diseases. In recent years, the sensing of intracellular DAMPs has received extensive attention in the field of sterile inflammation. However, emerging studies have shown that DAMPs that originate from neighbouring cells, and even from distal tissues or organs, also mediate sterile inflammatory responses. This multi-level sensing of DAMPs is crucial for intercellular, trans-tissue and trans-organ communication. Here, we summarize how DAMP-sensing receptors detect DAMPs from intracellular, intercellular or distal tissue and organ sources to mediate sterile inflammation. We also discuss the possibility of targeting DAMPs or their corresponding receptors to treat inflammatory diseases.
Collapse
Affiliation(s)
- Yi Huang
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Wei Jiang
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
9
|
Haroun E, Lim SH, Dutta D. GBT1118, a Voxelotor Analog, Ameliorates Hepatopathy in Sickle Cell Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1581. [PMID: 39459368 PMCID: PMC11509622 DOI: 10.3390/medicina60101581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: In sickle cell disease (SCD), hepatopathy is a cumulative consequence of ischemia/reperfusion (I/R) injury from a vaso-occlusive crisis, tissue inflammation, and iron overload due to blood transfusion. Hepatopathy is a major contributing factor of shortened life span in SCD patients. We hypothesized that the voxelotor, a hemoglobin allosteric modifier, ameliorates sickle hepatopathy. Materials and Methods: Townes SCD mice and their controls were treated with either chow containing GBT1118, a voxelotor analog, or normal chow. We evaluated inflammation, fibrosis, apoptosis and ferroptosis in their livers using qPCR, ELISA, histology, and immunohistochemistry. Results: GBT1118 treatment resulted in reduced hemolysis, iron overload and inflammation in the liver of SCD mice. There were significant reductions in the liver enzyme levels and bile acids. Furthermore, GBT1118-treated mice exhibited reduced apoptosis, necrosis, and fibrosis. Increased ferroptosis as evident from elevated 4-hydroxynonenal (4-HNE) staining, malondialdehyde (MDA) levels, and expression of Ptgs2 and Slc7a11 mRNAs, were also significantly reduced after GBT1118 treatment. To explain the increased ferroptosis, we evaluated iron homeostasis markers in livers. SCD mice showed decreased expression of heme oxygenase-1, ferritin, hepcidin, and ferroportin mRNA levels. GBT1118 treatment significantly increased expressions of these genes. Conclusions: Our results suggest GBT1118 treatment in SCD confers the amelioration of sickle hepatopathy by reducing inflammation, fibrosis, apoptosis, iron overload and ferroptosis.
Collapse
Affiliation(s)
| | | | - Dibyendu Dutta
- Division of Hematology and Oncology, Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (E.H.); (S.H.L.)
| |
Collapse
|
10
|
Wang W, Ma L, Liu B, Ouyang L. The role of trained immunity in sepsis. Front Immunol 2024; 15:1449986. [PMID: 39221248 PMCID: PMC11363069 DOI: 10.3389/fimmu.2024.1449986] [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: 06/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection, characterized by a systemic inflammatory response to infection. The use of antibiotics, fluid resuscitation, and organ support therapy has limited prognostic benefit in patients with sepsis, and its incidence is not diminishing, which is attracting increased attention in medicine. Sepsis remains one of the most debilitating and expensive illnesses. One of the main reasons of septic mortality is now understood to be disruption of immune homeostasis. Immunotherapy is revolutionizing the treatment of illnesses in which dysregulated immune responses play a significant role. This "trained immunity", which is a potent defense against infection regardless of the type of bacteria, fungus, or virus, is attributed to the discovery that the innate immune cells possess immune memory via metabolic and epigenetic reprogramming. Here we reviewed the immunotherapy of innate immune cells in sepsis, the features of trained immunity, and the relationship between trained immunity and sepsis.
Collapse
Affiliation(s)
| | | | | | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
11
|
Fu K, Zhao J, Zhong L, Xu H, Yu X, Bi X, Huang C. Dual therapy with phospholipase and metalloproteinase inhibitors from Sinonatrix annularis alleviated acute kidney and liver injury caused by multiple snake venoms. Biomed Pharmacother 2024; 177:116967. [PMID: 38908206 DOI: 10.1016/j.biopha.2024.116967] [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: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024] Open
Abstract
Snakebite envenomation often induces acute kidney injury (AKI) and acute liver injury (ALI), leading to augmented injuries and poor rehabilitation. Phospholipase A2 (PLA2) and metalloproteinase (SVMP) present in venom are responsible for the envenomation-associated events. In this study, mice envenomed with Deinagkistrodon acutus, Naja atra, or Agkistrodon halys pallas venom exhibited typical AKI and ALI symptoms, including significantly increased plasma levels of myoglobin, free hemoglobin, uric acid, aspartate aminotransferase, and alanine aminotransferase and upregulated expression of kidney NGAL and KIM-1. These effects were significantly inhibited when the mice were pretreated with natural inhibitors of PLA2 and SVMP isolated from Sinonatrix annularis (SaPLIγ and SaMPI). The inhibitors protected the physiological structural integrity of the renal tubules and glomeruli, alleviating inflammatory infiltration and diffuse hemorrhage in the liver. Furthermore, the dual therapy alleviated oxidative stress and apoptosis in the kidneys and liver by mitigating mitochondrial damage, thereby effectively reducing the lethal effect of snake venom in the inhibitor-treated mouse model. This study showed that dual therapy with inhibitors of metalloproteinase and phospholipase can effectively prevent ALI and AKI caused by snake bites. Our findings suggest that intrinsic inhibitors present in snakes are prospective therapeutic agents for multi-organ injuries caused by snake envenoming.
Collapse
Affiliation(s)
- Kepu Fu
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jianqi Zhao
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lipeng Zhong
- Clinical Laboratory Center, The First Affiliated Hospital, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330209, China
| | - Haiyan Xu
- Blood Transfusion Department, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
| | - Xinhui Yu
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaowen Bi
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chunhong Huang
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
12
|
Grujcic M, Milovanovic M, Nedeljkovic J, Jovanovic D, Arsenijevic D, Solovjova N, Stankovic V, Tanaskovic I, Arsenijevic A, Milovanovic J. The Possible Effects of Galectin-3 on Mechanisms of Renal and Hepatocellular Injury Induced by Intravascular Hemolysis. Int J Mol Sci 2024; 25:8129. [PMID: 39125698 PMCID: PMC11311984 DOI: 10.3390/ijms25158129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Intravascular hemolysis is a central feature of congenital and acquired hemolytic anemias, complement disorders, infectious diseases, and toxemias. Massive and/or chronic hemolysis is followed by the induction of inflammation, very often with severe damage of organs, which enhances the morbidity and mortality of hemolytic diseases. Galectin-3 (Gal-3) is a β-galactoside-binding lectin that modulates the functions of many immune cells, thus affecting inflammatory processes. Gal-3 is also one of the main regulators of fibrosis. The role of Gal-3 in the development of different kidney and liver diseases and the potential of therapeutic Gal-3 inhibition have been demonstrated. Therefore, the objective of this review is to discuss the possible effects of Gal-3 on the process of kidney and liver damage induced by intravascular hemolysis, as well as to shed light on the potential therapeutic targeting of Gal-3 in intravascular hemolysis.
Collapse
Affiliation(s)
- Mirjana Grujcic
- Institute for Transfusiology and Hemobiology of Military Medical Academy, 11000 Belgrade, Serbia;
| | - Marija Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Nedeljkovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danijela Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragana Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natalija Solovjova
- Academy of Applied Studies Belgrade, The College of Health Science, Cara Dušana 254, 11080 Belgrade, Serbia;
| | - Vesna Stankovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Irena Tanaskovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
13
|
Silveira THRE, Pereira DA, Pereira DA, Calmasini FB, Burnett AL, Costa FF, Silva FH. Impact of intravascular hemolysis on functional and molecular alterations in the urinary bladder: implications for an overactive bladder in sickle cell disease. Front Physiol 2024; 15:1369120. [PMID: 39100273 PMCID: PMC11294091 DOI: 10.3389/fphys.2024.1369120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/20/2024] [Indexed: 08/06/2024] Open
Abstract
Patients with sickle cell disease (SCD) display an overactive bladder (OAB). Intravascular hemolysis in SCD is associated with various severe SCD complications. However, no experimental studies have evaluated the effect of intravascular hemolysis on bladder function. This study aimed to assess the effects of intravascular hemolysis on the micturition process and the contractile mechanisms of the detrusor smooth muscle (DSM) in a mouse model with phenylhydrazine (PHZ)-induced hemolysis; furthermore, it aimed to investigate the role of intravascular hemolysis in the dysfunction of nitric oxide (NO) signaling and in increasing oxidative stress in the bladder. Mice underwent a void spot assay, and DSM contractions were evaluated in organ baths. The PHZ group exhibited increased urinary frequency and increased void volumes. DSM contractile responses to carbachol, KCl, α-β-methylene-ATP, and EFS were increased in the PHZ group. Protein expression of phosphorylated endothelial NO synthase (eNOS) (Ser-1177), phosphorylated neuronal NO synthase (nNOS) (Ser-1417), and phosphorylated vasodilator-stimulated phosphoprotein (VASP) (Ser-239) decreased in the bladder of the PHZ group. Protein expression of oxidative stress markers, NOX-2, 3-NT, and 4-HNE, increased in the bladder of the PHZ group. Our study shows that intravascular hemolysis promotes voiding dysfunction correlated with alterations in the NO signaling pathway in the bladder, as evidenced by reduced levels of p-eNOS (Ser-1177), nNOS (Ser-1417), and p-VASP (Ser-239). The study also showed that intravascular hemolysis increases oxidative stress in the bladder. Our study indicates that intravascular hemolysis promotes an OAB phenotype similar to those observed in patients and mice with SCD.
Collapse
Affiliation(s)
| | - Dalila Andrade Pereira
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, Brazil
| | - Danillo Andrade Pereira
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, Brazil
| | - Fabiano Beraldi Calmasini
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Arthur L. Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | | | - Fábio Henrique Silva
- Laboratory of Pharmacology, São Francisco University Medical School, Bragança Paulista, Brazil
| |
Collapse
|
14
|
Ben Soltana O, Barkallah M, Hentati F, Elhadef K, Ben Hlima H, Smaoui S, Michaud P, Abdelkafi S, Fendri I. Improving the shelf life of minced beef by Cystoseira compressa polysaccharide during storage. Int J Biol Macromol 2024; 273:132863. [PMID: 38838888 DOI: 10.1016/j.ijbiomac.2024.132863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
A polysaccharide extracted from the brown alga Cystoseira compressa (CCPS) was evaluated as a food additive to extend the shelf-life of raw beef meat. The antioxidant potential of CCPS was demonstrated by its inhibition of β-carotene bleaching (64.28 %), superoxide radicals (70.12 %), and hydroxyl radicals (93 %) at a concentration of 10 mg/ml. The polysaccharide also showed antibacterial activity with MIC values between 6.25 mg/ml and 50 mg/ml against five foodborne pathogenic bacteria. Furthermore, CCPS exhibited excellent functional, foaming, and emulsifying properties. Furthermore, microbiological and chemical effects of CCPS at concentrations equivalent to 1 MIC (CCPS-1), 2 MIC (CCPS-2), and 4 MIC (CCPS-3) were conducted. Chemical analyses showed that treated beef had significantly reduced TBARS levels below 2 mg MDA/kg at day 14. The treatment also decreased carbonyl groups, improved heme iron transformation, inhibited microbial growth (p < 0.05), and kept MetMb levels below 40 % by day 14. Moreover, two multivariate approaches, principal component analysis (PCA) and hierarchical cluster analysis (HCA), were effectively used to analyze the results characterizing the main attributes of the stored meat samples. In conclusion, these findings demonstrated that CCPS could be employed as a functional and bioactive component in the meat industry.
Collapse
Affiliation(s)
- Oumaima Ben Soltana
- Laboratoire de Génie Enzymatique et Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax 3038, Tunisia; Laboratoire de Biotechnologie des Plantes Appliquée à l'Amélioration des Cultures (LR01ES21), Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Mohamed Barkallah
- Laboratoire de Génie Enzymatique et Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax 3038, Tunisia
| | - Faiez Hentati
- Laboratoire de Génie Enzymatique et Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax 3038, Tunisia
| | - Khaoula Elhadef
- Laboratory of Microbial, Enzymatic Biotechnology and Biomolecules (LBMEB), Center of Biotechnology of Sfax, University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, 3018, Tunisia
| | - Hajer Ben Hlima
- Laboratoire de Génie Enzymatique et Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax 3038, Tunisia
| | - Slim Smaoui
- Laboratory of Microbial, Enzymatic Biotechnology and Biomolecules (LBMEB), Center of Biotechnology of Sfax, University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, 3018, Tunisia
| | - Philippe Michaud
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France
| | - Slim Abdelkafi
- Laboratoire de Génie Enzymatique et Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax 3038, Tunisia.
| | - Imen Fendri
- Laboratoire de Biotechnologie des Plantes Appliquée à l'Amélioration des Cultures (LR01ES21), Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| |
Collapse
|
15
|
Pradhan P, Vijayan V, Liu B, Martinez-Delgado B, Matamala N, Nikolin C, Greite R, DeLuca DS, Janciauskiene S, Motterlini R, Foresti R, Immenschuh S. Distinct metabolic responses to heme in inflammatory human and mouse macrophages - Role of nitric oxide. Redox Biol 2024; 73:103191. [PMID: 38762951 PMCID: PMC11130737 DOI: 10.1016/j.redox.2024.103191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024] Open
Abstract
Activation of inflammation is tightly associated with metabolic reprogramming in macrophages. The iron-containing tetrapyrrole heme can induce pro-oxidant and pro-inflammatory effects in murine macrophages, but has been associated with polarization towards an anti-inflammatory phenotype in human macrophages. In the current study, we compared the regulatory responses to heme and the prototypical Toll-like receptor (TLR)4 ligand lipopolysaccharide (LPS) in human and mouse macrophages with a particular focus on alterations of cellular bioenergetics. In human macrophages, bulk RNA-sequencing analysis indicated that heme led to an anti-inflammatory transcriptional profile, whereas LPS induced a classical pro-inflammatory gene response. Co-stimulation of heme with LPS caused opposing regulatory patterns of inflammatory activation and cellular bioenergetics in human and mouse macrophages. Specifically, in LPS-stimulated murine, but not human macrophages, heme led to a marked suppression of oxidative phosphorylation and an up-regulation of glycolysis. The species-specific alterations in cellular bioenergetics and inflammatory responses to heme were critically dependent on the availability of nitric oxide (NO) that is generated in inflammatory mouse, but not human macrophages. Accordingly, studies with an inducible nitric oxide synthase (iNOS) inhibitor in mouse, and a pharmacological NO donor in human macrophages, reveal that NO is responsible for the opposing effects of heme in these cells. Taken together, the current findings indicate that NO is critical for the immunomodulatory role of heme in macrophages.
Collapse
Affiliation(s)
- Pooja Pradhan
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Vijith Vijayan
- Department of Pediatrics, Stanford University, Stanford, USA
| | - Bin Liu
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Beatriz Martinez-Delgado
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220, Madrid, Spain
| | - Nerea Matamala
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220, Madrid, Spain
| | - Christoph Nikolin
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Robert Greite
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - David S. DeLuca
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | | | - Roberta Foresti
- University Paris-Est Créteil, INSERM, IMRB, F-94010, Créteil, France
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, Aschner M, Tizabi Y. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol 2024; 7:100181. [PMID: 39021403 PMCID: PMC11252799 DOI: 10.1016/j.crtox.2024.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobin disorder marked by red blood cell sickling, resulting in severe anemia, painful episodes, extensive organ damage, and shortened life expectancy. In SCD, increased iron levels can trigger ferroptosis, a specific type of cell death characterized by reactive oxygen species (ROS) and lipid peroxide accumulation, leading to damage and organ impairments. The intricate interplay between iron, ferroptosis, inflammation, and oxidative stress in SCD underscores the necessity of thoroughly understanding these processes for the development of innovative therapeutic strategies. This review highlights the importance of balancing the complex interactions among various factors and exploitation of the knowledge in developing novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Jaqueline Lima
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Gabriel F. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Yasmin S. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Departments of Microbiology and Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
17
|
Shah SS, Stone EF, Francis RO, Karafin MS. The global role of G6PD in infection and immunity. Front Immunol 2024; 15:1393213. [PMID: 38938571 PMCID: PMC11208698 DOI: 10.3389/fimmu.2024.1393213] [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: 03/04/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is the most common enzymopathy in humans. G6PD is an essential enzyme in the pentose phosphate pathway (PPP), generating NADPH needed for cellular biosynthesis and reactive oxygen species (ROS) homeostasis, the latter especially key in red blood cells (RBCs). Beyond the RBC, there is emerging evidence that G6PD exerts an immunologic role by virtue of its functions in leukocyte oxidative metabolism and anabolic synthesis necessary for immune effector function. We review these here, and consider the global immunometabolic role of G6PD activity and G6PD deficiency in modulating inflammation and immunopathology.
Collapse
Affiliation(s)
- Shivang S. Shah
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Elizabeth F. Stone
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Richard O. Francis
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Matthew S. Karafin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
18
|
de Ligt LA, Gaartman AE, Biemond BJ, Fijnvandraat K, van Bruggen R, Nur E. Neutrophils in sickle cell disease: Exploring their potential role as a therapeutic target. Am J Hematol 2024; 99:1119-1128. [PMID: 38293835 DOI: 10.1002/ajh.27224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/15/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024]
Abstract
Factors influencing the activation of neutrophils in SCD and the potential neutrophil-mediated ameliorating effects of therapies in SCD.
Collapse
Affiliation(s)
- Lydian A de Ligt
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Aafke E Gaartman
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Bart J Biemond
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Erfan Nur
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Kostelac D, Dolenec F, Markovinović AB, Markov K, Bursać Kovačević D, Frece J. The Application of Probiotic Bacteria from Strawberry ( Fragaria ananassa × Duch.) in the Fermentation of Strawberry Tree Fruit ( Arbutus unedo L.) Extract. Microorganisms 2024; 12:1000. [PMID: 38792829 PMCID: PMC11124331 DOI: 10.3390/microorganisms12051000] [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: 02/05/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The search for unexplored plant resources that would provide a good basis for the development of novel probiotic functional foods is rapidly increasing. In this context, the strawberry tree fruit (Arbutus unedo L.) is particularly interesting, as it is rich in numerous antioxidant bioactive compounds that have been shown to be beneficial to health, but have not yet found industrial applications. In this work, the probiotic characterization of lactic acid bacteria strain Lactiplantibacillus plantarum DB2, isolated from strawberries (Fragaria ananassa × Duch.), was performed. The tested strain proved to be safe to use, displaying no antibiotic resistance or hemolytic activity. Due to its proven probiotic potential during simulated gastrointestinal transit, its antimicrobial activity, and its coaggregation with pathogens, it was selected for fermentation of an aqueous Arbutus unedo L. extract, which was subsequently microencapsulated and freeze-dried to extend its shelf life and preserve its functional properties. The antioxidant activity of the ferment obtained was maintained (80%), while after microencapsulation and freeze-drying, about 50% and 20% of the antioxidant activity was retained, respectively. In conclusion, this study demonstrates for the first time the application of probiotics isolated from strawberries in the fermentation of strawberry tree extract and monitors the antioxidant activity during post-fermentation formulation, paving the way for a potential industrial application of this underutilized plant.
Collapse
Affiliation(s)
| | | | | | | | | | - Jadranka Frece
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (D.K.); (F.D.); (A.B.M.); (K.M.); (D.B.K.)
| |
Collapse
|
20
|
Brattinga B, Plas M, Spikman JM, Rutgers A, de Haan JJ, van der Wal-Huisman H, Absalom AR, Nieuwenhuijs-Moeke GJ, van Munster BC, de Bock GH, van Leeuwen BL. The link between the early surgery-induced inflammatory response and postoperative cognitive dysfunction in older patients. J Am Geriatr Soc 2024; 72:1360-1372. [PMID: 38516716 DOI: 10.1111/jgs.18876] [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: 05/03/2023] [Revised: 01/14/2024] [Accepted: 02/25/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a common complication in older patients with cancer and is associated with decreased quality of life and increased disability and mortality rates. Systemic inflammation resulting in neuroinflammation is considered important in the pathogenesis of POCD. The aim of this study was to explore the association between the early surgery-induced inflammatory response and POCD within 3 months after surgery in older cancer patients. METHODS Patients ≥65 years in need of surgery for a solid tumor were included in a prospective cohort study. Plasma levels of C-reactive protein (CRP), interleukin-1 beta (IL-1β), IL-6, IL-10, and Neutrophil gelatinase-associated lipocalin (NGAL) were measured perioperatively. Cognitive performance was assessed preoperatively and 3 months after surgery. POCD was defined as a decline in cognitive test scores of ≥25% on ≥2 of five tests within the different cognitive domains of memory, executive functioning, and information processing speed. Logistic regression analysis was performed. RESULTS POCD was observed in 44 (17.7%) of 248 included patients. Age >75, preoperative Mini-Mental State Examination (MMSE) score ≤26 and major surgery were independent significant predictors for POCD. In multivariate logistic regression analysis, no significant associations were shown between the early surgery-induced inflammatory response and either POCD or decline within the different cognitive domains. CONCLUSIONS This study shows that one out of six older patients with cancer developed POCD within 3 months after surgery. The early surgery-induced inflammatory response was neither associated with POCD, nor with decline in the separate cognitive domains. Further research is necessary for better understanding of the complex etiology of POCD.
Collapse
Affiliation(s)
- Baukje Brattinga
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Matthijs Plas
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacoba M Spikman
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, University Medical Center, Groningen, The Netherlands
| | - Jacco J de Haan
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Anthony R Absalom
- Department of Anesthesiology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Barbara C van Munster
- Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara L van Leeuwen
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
22
|
Du Y, Lu C, Bi L, Wang C, Zhao M, Ding Y, Fan W. Causal effects of genetically determined metabolites on androgenetic alopecia: A two-sample Mendelian randomization analysis. Skin Res Technol 2024; 30:e13732. [PMID: 38747971 PMCID: PMC11095465 DOI: 10.1111/srt.13732] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/01/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Androgenic alopecia (AGA) is the most common non-scarring alopecia disorder. Given its increasing incidence and onset during adolescence, AGA significantly impacts both the physical and psychological well-being of affected individuals. Emerging evidence suggests a pivotal role of metabolites in AGA. This study aims to elucidate the causal relationship between metabolites and AGA using Mendelian randomization (MR) analysis. METHODS We conducted a two-sample Mendelian randomization (TSMR) analysis based on a genome-wide association study (GWAS) to assess the causality of 452 metabolites on AGA. The main approach employed for inferring causal effects was inverse variance weighted (IVW), which was complemented by MR-Egger regression, weighted median, as well as MR pleiotropy residual sum and outlier (MR-PRESSO) approaches. Additionally, sensitivity analyses were performed to ensure result robustness. Single nucleotide polymorphisms (SNPs) were selected as instrumental variables (IVs) in GWAS dataset comprising 452 metabolites. RESULTS Notably, we identified Scyllo-inositol and Alpha-ketoglutarate as the most potent protective factors against AGA, while Heme* and 2-palmitoylglycerophosphocholine* emerged as significant risk factors for AGA. Furthermore, sensitivity analysis revealed no heterogeneity in these findings. CONCLUSIONS Overall, our research suggests a potential causal link between metabolites and AGA, offering a more comprehensive insight into the pathogenesis of AGA and present additional strategies for prevention and treatment.
Collapse
Affiliation(s)
- Yimei Du
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Changpei Lu
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Lingbo Bi
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Chaofan Wang
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Min Zhao
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Yunbu Ding
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Weixin Fan
- Department of DermatologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| |
Collapse
|
23
|
Eckhart L, Fischer H. Caspase-5: Structure, Pro-Inflammatory Activity and Evolution. Biomolecules 2024; 14:520. [PMID: 38785927 PMCID: PMC11117641 DOI: 10.3390/biom14050520] [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: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Caspase-5 is a protease that induces inflammation in response to lipopolysaccharide (LPS), a component of the cell envelope of Gram-negative bacteria. The expression level of the CASP5 gene is very low in the basal state, but strongly increases in the presence of LPS. Intracellular LPS binds to the caspase activation and recruitment domain (CARD) of caspase-5, leading to the formation of a non-canonical inflammasome. Subsequently, the catalytic domain of caspase-5 cleaves gasdermin D and thereby facilitates the formation of cell membrane pores through which pro-inflammatory cytokines of the interleukin-1 family are released. Caspase-4 is also able to form a non-canonical inflammasome upon binding to LPS, but its expression is less dependent on LPS than the expression of caspase-5. Caspase-4 and caspase-5 have evolved via the duplication of a single ancestral gene in a subclade of primates, including humans. Notably, the main biomedical model species, the mouse, has only one ortholog, namely caspase-11. Here, we review the structural features and the mechanisms of regulation that are important for the pro-inflammatory roles of caspase-5. We summarize the interspecies differences and the evolution of pro-inflammatory caspases in mammals and discuss the potential roles of caspase-5 in the defense against Gram-negative bacteria and in sepsis.
Collapse
Affiliation(s)
- Leopold Eckhart
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Heinz Fischer
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
24
|
Kasper R, Rodriguez-Alfonso A, Ständker L, Wiese S, Schneider EM. Major endothelial damage markers identified from hemadsorption filters derived from treated patients with septic shock - endoplasmic reticulum stress and bikunin may play a role. Front Immunol 2024; 15:1359097. [PMID: 38698864 PMCID: PMC11063272 DOI: 10.3389/fimmu.2024.1359097] [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: 12/20/2023] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction In septic patients the damage of the endothelial barrier is decisive leading to circulatory septic shock with disseminated vascular coagulation, edema and multiorgan failure. Hemadsorption therapy leads to rapid resolution of clinical symptoms. We propose that the isolation of proteins adsorbed to hemadsorption devices contributes to the identification of mediators responsible for endothelial barrier dysfunction. Material and methods Plasma materials enriched to hemadsorption filters (CytoSorb®) after therapy of patients in septic shock were fractionated and functionally characterized for their effect on cell integrity, viability, proliferation and ROS formation by human endothelial cells. Fractions were further studied for their contents of oxidized nucleic acids as well as peptides and proteins by mass spectrometry. Results Individual fractions exhibited a strong effect on endothelial cell viability, the endothelial layer morphology, and ROS formation. Fractions with high amounts of DNA and oxidized DNA correlated with ROS formation in the target endothelium. In addition, defined proteins such as defensins (HNP-1), SAA1, CXCL7, and the peptide bikunin were linked to the strongest additive effects in endothelial damage. Conclusion Our results indicate that hemadsorption is efficient to transiently remove strong endothelial damage mediators from the blood of patients with septic shock, which explains a rapid clinical improvement of inflammation and endothelial function. The current work indicates that a combination of stressors leads to the most detrimental effects. Oxidized ssDNA, likely derived from mitochondria, SAA1, the chemokine CXCL7 and the human neutrophil peptide alpha-defensin 1 (HNP-1) were unique for their significant negative effect on endothelial cell viability. However, the strongest damage effect occurred, when, bikunin - cleaved off from alpha-1-microglobulin was present in high relative amounts (>65%) of protein contents in the most active fraction. Thus, a relevant combination of stressors appears to be removed by hemadsorption therapy which results in fulminant and rapid, though only transient, clinical restitution.
Collapse
Affiliation(s)
- Robin Kasper
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Armando Rodriguez-Alfonso
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - E. Marion Schneider
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
25
|
Yeudall S, Upchurch CM, Leitinger N. The clinical relevance of heme detoxification by the macrophage heme oxygenase system. Front Immunol 2024; 15:1379967. [PMID: 38585264 PMCID: PMC10995405 DOI: 10.3389/fimmu.2024.1379967] [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: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Heme degradation by the heme oxygenase (HMOX) family of enzymes is critical for maintaining homeostasis and limiting heme-induced tissue damage. Macrophages express HMOX1 and 2 and are critical sites of heme degradation in healthy and diseased states. Here we review the functions of the macrophage heme oxygenase system and its clinical relevance in discrete groups of pathologies where heme has been demonstrated to play a driving role. HMOX1 function in macrophages is essential for limiting oxidative tissue damage in both acute and chronic hemolytic disorders. By degrading pro-inflammatory heme and releasing anti-inflammatory molecules such as carbon monoxide, HMOX1 fine-tunes the acute inflammatory response with consequences for disorders of hyperinflammation such as sepsis. We then discuss divergent beneficial and pathological roles for HMOX1 in disorders such as atherosclerosis and metabolic syndrome, where activation of the HMOX system sits at the crossroads of chronic low-grade inflammation and oxidative stress. Finally, we highlight the emerging role for HMOX1 in regulating macrophage cell death via the iron- and oxidation-dependent form of cell death, ferroptosis. In summary, the importance of heme clearance by macrophages is an active area of investigation with relevance for therapeutic intervention in a diverse array of human diseases.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Clint M. Upchurch
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
26
|
Nascimento Júnior JXD, Sola-Penna M, Zancan P. Clotrimazole reverses macrophage M2 polarization by disrupting the PI3K/AKT/mTOR pathway. Biochem Biophys Res Commun 2024; 696:149455. [PMID: 38176247 DOI: 10.1016/j.bbrc.2023.149455] [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: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Macrophages switch among different activation phenotypes according to distinct environmental stimuli, varying from pro-inflammatory (M1) to alternative (also named resolutive; M2) activation forms. M1-and M2-activated macrophages represent the two extremes of the activation spectrum involving multiple species, which vary in terms of function and the cytokines secreted. The consensus is that molecular characterization of the distinct macrophage population and the signals driving their activation will help in explaining disease etiology and formulating therapies. For instance, myeloid cells residing in the tumor microenvironment are key players in tumor progression and usually display an M2-like phenotype, which help tumor cells to evade local inflammatory processes. Therefore, these specific cells have been proposed as targets for tumor therapies by changing their activation profile. Furthermore, M2 polarized macrophages are phagocytic cells promoting tissue repair and wound healing and are therefore potential targets to treat different diseases. We have already shown that clotrimazole (CTZ) decreases tumor cell viability and thus tumor growth. The mechanism by which CTZ exerts its effects remains to be determined, but this drug is an inhibitor of the PI3K/AKT/mTOR pathway. In this study, we show that CTZ downregulated M2-activation markers in macrophages polarized to the M2 profile. This effect occurred without interfering with the expression of M1-polarized markers or pro-inflammatory cytokines and signaling. Moreover, CTZ suppressed NFkB pathway intermediates and disrupted PI3K/AKT/mTOR signaling. We concluded that CTZ reverses macrophage M2 polarization by disrupting the PI3K/AKT/mTOR pathway, which results in the suppression of NFkB induction of M2 polarization. In addition, we find that CTZ represents a promising therapeutic tool as an antitumor agent.
Collapse
Affiliation(s)
- José Xavier do Nascimento Júnior
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Mauro Sola-Penna
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Patricia Zancan
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
27
|
Hopp MT, Holze J, Lauber F, Holtkamp L, Rathod DC, Miteva MA, Prestes EB, Geyer M, Manoury B, Merle NS, Roumenina LT, Bozza MT, Weindl G, Imhof D. Insights into the molecular basis and mechanism of heme-triggered TLR4 signalling: The role of heme-binding motifs in TLR4 and MD2. Immunology 2024; 171:181-197. [PMID: 37885279 DOI: 10.1111/imm.13708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Haemolytic disorders, such as sickle cell disease, are accompanied by the release of high amounts of labile heme into the intravascular compartment resulting in the induction of proinflammatory and prothrombotic complications in affected patients. In addition to the relevance of heme-regulated proteins from the complement and blood coagulation systems, activation of the TLR4 signalling pathway by heme was ascribed a crucial role in the progression of these pathological processes. Heme binding to the TLR4-MD2 complex has been proposed recently, however, essential mechanistic information of the processes at the molecular level, such as heme-binding kinetics, the heme-binding capacity and the respective heme-binding sites (HBMs) is still missing. We report the interaction of TLR4, MD2 and the TLR4-MD2 complex with heme and the consequences thereof by employing biochemical, spectroscopic, bioinformatic and physiologically relevant approaches. Heme binding occurs transiently through interaction with up to four HBMs in TLR4, two HBMs in MD2 and at least four HBMs in their complex. Functional studies highlight that mutations of individual HBMs in TLR4 preserve full receptor activation by heme, suggesting that heme interacts with TLR4 through different binding sites independently of MD2. Furthermore, we confirm and extend the major role of TLR4 for heme-mediated cytokine responses in human immune cells.
Collapse
Affiliation(s)
- Marie-T Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany
- Department of Chemistry, Institute of Integrated Natural Sciences, University of Koblenz, Koblenz, Germany
| | - Janine Holze
- Pharmacology and Toxicology, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Felicitas Lauber
- Pharmacology and Toxicology, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Laura Holtkamp
- Pharmacology and Toxicology, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Dhruv C Rathod
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Maria A Miteva
- CNRS UMR 8038 CiTCoM, Université de Paris, Faculté de Pharmacie de Paris, Paris, France
- INSERM U 1268 Medicinal Chemistry and Translational Research, Paris, France
| | - Elisa B Prestes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR8253, Université Paris Cité, Faculté de médecine Necker, Paris, France
| | - Nicolas S Merle
- Centre de Recherche des Cordeliers, UMR_S 1138, INSERM, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Universités, Paris, France
- Centre de Recherche des Cordeliers, Université Paris Descartes, Paris, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, UMR_S 1138, INSERM, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Universités, Paris, France
- Centre de Recherche des Cordeliers, Université Paris Descartes, Paris, France
| | - Marcelo T Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Günther Weindl
- Pharmacology and Toxicology, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
28
|
Shete S, Iqbal F, Bhardwaj M, Nandi U, Kumar A, Reddy DS. Sila-CBD Derivatives as Inhibitors of Heme-Induced NLRP3 Inflammasome: Application in Hemolytic Diseases. ACS Med Chem Lett 2023; 14:1716-1723. [PMID: 38116428 PMCID: PMC10726456 DOI: 10.1021/acsmedchemlett.3c00352] [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: 08/08/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 12/21/2023] Open
Abstract
Synthesis and biological evaluation of silicon-incorporated phytocannabinoids with improved pharmacological properties toward inflammatory diseases are described. The synthesized sila-analogues 15a, 15b, and 15c displayed potent inhibition of pro-inflammatory cytokines, including IL-1β, TNF-α, and IL-6 at 10 μM. Further, the release of heme during the lysis of red blood cells in hemolytic diseases is one of the major reasons for inflammation associated with the pathophysiology of these diseases. Due to scanty literature related to inhibitors of heme-mediated induction of the NLRP3 inflammasome, we decided to test these compounds against it. Compounds 15a and 15c significantly inhibited the heme-mediated induction of the NLRP3 inflammasome at a concentration of 0.1 μM. Interestingly, the sila-CBD derivatives also showed higher metabolic stability in contrast to their carbon analogues. Anti-NLRP3 inflammasome activity of compounds 15a and 15c were further validated in vivo against heme-mediated peritoneal inflammation. The anti-inflammatory activity of these compounds could be useful in treating diseases such as sickle cell anemia and thalassemia involving the hemolysis-mediated activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Sanket
S. Shete
- Division
of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
- Natural
Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Fiza Iqbal
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Mahir Bhardwaj
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Utpal Nandi
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - Ajay Kumar
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Canal Road, Jammu 180001, India
| | - D. Srinivasa Reddy
- Division
of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
- Natural
Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
29
|
Amaral MP, Cardoso FD, de Farias IS, de Souza RQ, Matteucci KC, Torrecilhas AC, Bortoluci KR. NAIP/NLRC4 inflammasome participates in macrophage responses to Trypanosoma cruzi by a mechanism that relies on cathepsin-dependent caspase-1 cleavage. Front Immunol 2023; 14:1282856. [PMID: 38124741 PMCID: PMC10731265 DOI: 10.3389/fimmu.2023.1282856] [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: 08/24/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Inflammasomes are large protein complexes that, once activated, initiate inflammatory responses by activating the caspase-1 protease. They play pivotal roles in host defense against pathogens. The well-established role of NAIP/NLRC4 inflammasome in bacterial infections involves NAIP proteins functioning as sensors for their ligands. However, recent reports have indicated the involvement of NLRC4 in non-bacterial infections and sterile inflammation, even though the role of NAIP proteins and the exact molecular mechanisms underlying inflammasome activation in these contexts remain to be elucidated. In this study, we investigated the activation of the NAIP/NLRC4 inflammasome in response to Trypanosoma cruzi, the protozoan parasite responsible for causing Chagas disease. This parasite has been previously demonstrated to activate NLRP3 inflammasomes. Here we found that NAIP and NLRC4 proteins are also required for IL-1β and Nitric Oxide (NO) release in response to T. cruzi infection, with their absence rendering macrophages permissive to parasite replication. Moreover, Nlrc4 -/- and Nlrp3 -/- macrophages presented similar impaired responses to T. cruzi, underscoring the non-redundant roles played by these inflammasomes during infection. Notably, it was the live trypomastigotes rather than soluble antigens or extracellular vesicles (EVs) secreted by them, that activated inflammasomes in a cathepsins-dependent manner. The inhibition of cathepsins effectively abrogated caspase-1 cleavage, IL-1β and NO release, mirroring the phenotype observed in Nlrc4 -/-/Nlrp3 -/- double knockout macrophages. Collectively, our findings shed light on the pivotal role of the NAIP/NLRC4 inflammasome in macrophage responses to T. cruzi infection, providing new insights into its broader functions that extend beyond bacterial infections.
Collapse
Affiliation(s)
- Marcelo Pires Amaral
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Felipe Daniel Cardoso
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Ingrid Sancho de Farias
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Rafael Queiroz de Souza
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Kely Catarine Matteucci
- Plataforma de Medicina Translacional, Fundação Oswaldo Cruz (FIOCRUZ), Faculdade de Medicina de Ribeirão Preto (FMRP), Ribeirão Preto, SP, Brazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Karina Ramalho Bortoluci
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
30
|
Othman B, Zeef L, Szestak T, Rchiad Z, Storm J, Askonas C, Satyam R, Madkhali A, Haley M, Wagstaff S, Couper K, Pain A, Craig A. Different PfEMP1-expressing Plasmodium falciparum variants induce divergent endothelial transcriptional responses during co-culture. PLoS One 2023; 18:e0295053. [PMID: 38033133 PMCID: PMC10688957 DOI: 10.1371/journal.pone.0295053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
The human malaria parasite Plasmodium falciparum is responsible for the majority of mortality and morbidity caused by malaria infection and differs from other human malaria species in the degree of accumulation of parasite-infected red blood cells in the microvasculature, known as cytoadherence or sequestration. In P. falciparum, cytoadherence is mediated by a protein called PfEMP1 which, due to its exposure to the host immune system, undergoes antigenic variation resulting in the expression of different PfEMP1 variants on the infected erythrocyte membrane. These PfEMP1s contain various combinations of adhesive domains, which allow for the differential engagement of a repertoire of endothelial receptors on the host microvasculature, with specific receptor usage associated with severe disease. We used a co-culture model of cytoadherence incubating human brain microvascular endothelial cells with erythrocytes infected with two parasite lines expressing different PfEMP1s that demonstrate different binding profiles to vascular endothelium. We determined the transcriptional profile of human brain microvascular endothelial cells (HBMEC) following different incubation periods with infected erythrocytes, identifying different transcriptional profiles of pathways previously found to be involved in the pathology of severe malaria, such as inflammation, apoptosis and barrier integrity, induced by the two PfEMP1 variants.
Collapse
Affiliation(s)
- Basim Othman
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Leo Zeef
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tadge Szestak
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Zineb Rchiad
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Janet Storm
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Caroline Askonas
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Rohit Satyam
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Aymen Madkhali
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Michael Haley
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Simon Wagstaff
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Kevin Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Arnab Pain
- Pathogen Genomics Laboratory, Bioscience Program, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal, KSA
| | - Alister Craig
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| |
Collapse
|
31
|
Tweedell RE, Kanneganti T. NLRP12-PANoptosome in haemolytic, infectious and inflammatory diseases. Clin Transl Med 2023; 13:e1409. [PMID: 37700491 PMCID: PMC10497829 DOI: 10.1002/ctm2.1409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Affiliation(s)
- Rebecca E. Tweedell
- Department of ImmunologySt. Jude Children's Research HospitalMemphisTennesseeUSA
| | | |
Collapse
|
32
|
Kahan R, Cray PL, Abraham N, Gao Q, Hartwig MG, Pollara JJ, Barbas AS. Sterile inflammation in liver transplantation. Front Med (Lausanne) 2023; 10:1223224. [PMID: 37636574 PMCID: PMC10449546 DOI: 10.3389/fmed.2023.1223224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Sterile inflammation is the immune response to damage-associated molecular patterns (DAMPs) released during cell death in the absence of foreign pathogens. In the setting of solid organ transplantation, ischemia-reperfusion injury results in mitochondria-mediated production of reactive oxygen and nitrogen species that are a major cause of uncontrolled cell death and release of various DAMPs from the graft tissue. When properly regulated, the immune response initiated by DAMP-sensing serves as means of damage control and is necessary for initiation of recovery pathways and re-establishment of homeostasis. In contrast, a dysregulated or overt sterile inflammatory response can inadvertently lead to further injury through recruitment of immune cells, innate immune cell activation, and sensitization of the adaptive immune system. In liver transplantation, sterile inflammation may manifest as early graft dysfunction, acute graft failure, or increased risk of immunosuppression-resistant rejection. Understanding the mechanisms of the development of sterile inflammation in the setting of liver transplantation is crucial for finding reliable biomarkers that predict graft function, and for development of therapeutic approaches to improve long-term transplant outcomes. Here, we discuss the recent advances that have been made to elucidate the early signs of sterile inflammation and extent of damage from it. We also discuss new therapeutics that may be effective in quelling the detrimental effects of sterile inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrew S. Barbas
- Duke Ex-Vivo Organ Lab (DEVOL)—Division of Abdominal Transplant Surgery, Duke University, Durham, NC, United States
| |
Collapse
|
33
|
LIU D, ZHANG Y, YU T, LIU Z, JIAO Y, WANG H, XU Y, GUAN Q, CHEN L, HU H. Protective mechanisms of Tuina therapy against lipopolysaccharide-induced fever in young rabbits based on untargeted metabolomics analysis. J TRADIT CHIN MED 2023; 43:725-733. [PMID: 37454257 PMCID: PMC10320445 DOI: 10.19852/j.cnki.jtcm.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/15/2022] [Indexed: 07/18/2023]
Abstract
OBJECTIVE To investigate the effect of Tuina on the plasma metabolites of lipopolysaccharide-induced febrile in infant rabbits. METHODS Twenty-four infant New Zealand rabbits were selected and randomly divided into three groups: saline, model, and Tuina. The fever model was established by injecting LPS intravenously through the ear margin vein in the model group and Tuina group, respectively. The modeling was considered successful when the anal temperature increased by 0.5℃ or above within 1 h. In the Tuina group, six Tuina techniques (i.e., opening Tianmen / the heaven gate, pushing Kangong / the superciliary arch, kneading Taiyang and the prominent bone behind the ears, clearing Tianheshui, spine pinching) that alleviate fever were performed on the young rabbits 1 h after the modeling, whereas the model and saline groups were not given Tuina treatment, with the real-time anal temperature monitored during the experiment. The plasma was taken 3 h after the modeling for liquid chromatography-mass spectrometry (LC-MS) untargeted metabolomics study. RESULTS Our results showed a fever-reducing effects of Tuina therapy on lipopolysaccharide-induced fever in young rabbits, as indicated by a significantly lower anal temperature, maximum rise in body temperature, and body response index at 2 and 3 h after modeling in the Tuina group compared to the model group, with reductions in the PGE2 expression observed in the blood and hypothalamus. The differential metabolites including riboflavin, nicotinamide N-oxide, porphobilinogen, 5-hydroxyindoleacetic acid, gamma-aminobutyric acid, and lysoPC (16:1 (9Z)/0:0) were found following the Tuina intervention. Tuina primarily involves glycine-serine-threonine, arginine-proline, porphyrin-chlorophyll, pyrimidine, primary bile acid biosynthesis, and cyanoamino acid metabolic pathways. CONCLUSION Tuina therapy has proven to be effective in reducing body temperature and down-regulating PGE2 expression in LPS-induced febrile young rabbits, with its mechanism of fever-reducing action possibly associated with the changes in plasma metabolites and metabolic pathways.
Collapse
Affiliation(s)
- Di LIU
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Yingqi ZHANG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Tianyuan YU
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Zhifeng LIU
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Yi JIAO
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Hourong WANG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Yajing XU
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Qian GUAN
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Lulu CHEN
- 2 Department of Acupuncture-Moxibustion, Beijing Massage Hospital, Beijing 100035, China
| | - Hui HU
- 3 Department of Acupuncture-Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| |
Collapse
|
34
|
de Lima F, Hounkpe BW, de Moraes CRP, Borba-Junior IT, Costa FF, De Paula EV. Safety and feasibility of the gene transfer of hemopexin for conditions with increased free heme. Exp Biol Med (Maywood) 2023; 248:1103-1111. [PMID: 37452705 PMCID: PMC10583756 DOI: 10.1177/15353702231182199] [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: 01/30/2023] [Accepted: 03/16/2023] [Indexed: 07/18/2023] Open
Abstract
Heme is a fundamental molecule for several biological processes, but when released in the extracellular space such as in hemolytic diseases, it can be toxic to cells and tissues. Hemopexin (HPX) is a circulating protein responsible for removing free heme from the circulation, whose levels can be severely depleted in conditions such as sickle cell diseases. Accordingly, increasing HPX levels represents an attractive strategy to mitigate the deleterious effects of heme in these conditions. Gene transfer of liver-produced proteins with adeno-associated virus (AAV) has been shown to be an effective and safety strategy in animal and human studies mainly in hemophilia. Here, we report the feasibility of increasing HPX levels using an AAV8 vector expressing human HPX (hHPX). C57Bl mice were injected with escalating doses of our vector, and expression was assessed by enzyme immunoassay (ELISA), Western blot, and quantitative polymerase chain reaction (qPCR). In addition, the biological activity of transgenic hHPX was confirmed using two different models of heme challenge consisting of serial heme injections or phenylhydrazine-induced hemolysis. Sustained expression of hHPX was confirmed for up to 26 weeks in plasma. Expression was dose-dependent and not associated with clinical signs of toxicity. hHPX levels were significantly reduced by heme infusions and phenylhydrazine-induced hemolysis. No clinical toxicity or laboratory signs of liver damage were observed in preliminary short-term heme challenge studies. Our results confirm that long-term expression of hHPX is feasible and safe in mice, even in the presence of heme overload. Additional studies are needed to explore the effect of transgenic HPX protein in animal models of chronic hemolysis.
Collapse
Affiliation(s)
- Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | | | | | - Fernando Ferreira Costa
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| | - Erich V De Paula
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| |
Collapse
|
35
|
Sundaram B, Pandian N, Mall R, Wang Y, Sarkar R, Kim HJ, Malireddi RKS, Karki R, Janke LJ, Vogel P, Kanneganti TD. NLRP12-PANoptosome activates PANoptosis and pathology in response to heme and PAMPs. Cell 2023; 186:2783-2801.e20. [PMID: 37267949 PMCID: PMC10330523 DOI: 10.1016/j.cell.2023.05.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/17/2023] [Accepted: 05/05/2023] [Indexed: 06/04/2023]
Abstract
Cytosolic innate immune sensors are critical for host defense and form complexes, such as inflammasomes and PANoptosomes, that induce inflammatory cell death. The sensor NLRP12 is associated with infectious and inflammatory diseases, but its activating triggers and roles in cell death and inflammation remain unclear. Here, we discovered that NLRP12 drives inflammasome and PANoptosome activation, cell death, and inflammation in response to heme plus PAMPs or TNF. TLR2/4-mediated signaling through IRF1 induced Nlrp12 expression, which led to inflammasome formation to induce maturation of IL-1β and IL-18. The inflammasome also served as an integral component of a larger NLRP12-PANoptosome that drove inflammatory cell death through caspase-8/RIPK3. Deletion of Nlrp12 protected mice from acute kidney injury and lethality in a hemolytic model. Overall, we identified NLRP12 as an essential cytosolic sensor for heme plus PAMPs-mediated PANoptosis, inflammation, and pathology, suggesting that NLRP12 and molecules in this pathway are potential drug targets for hemolytic and inflammatory diseases.
Collapse
Affiliation(s)
- Balamurugan Sundaram
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nagakannan Pandian
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yaqiu Wang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Roman Sarkar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hee Jin Kim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Laura J Janke
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
36
|
Chatterjee T, Arora I, Underwood LB, Lewis TL, Masjoan Juncos JX, Heath SL, Goodin BR, Aggarwal S. Heme-Induced Macrophage Phenotype Switching and Impaired Endogenous Opioid Homeostasis Correlate with Chronic Widespread Pain in HIV. Cells 2023; 12:1565. [PMID: 37371035 PMCID: PMC10297192 DOI: 10.3390/cells12121565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic widespread pain (CWP) is associated with a high rate of disability and decreased quality of life in people with HIV-1 (PWH). We previously showed that PWH with CWP have increased hemolysis and elevated plasma levels of cell-free heme, which correlate with low endogenous opioid levels in leukocytes. Further, we demonstrated that cell-free heme impairs β-endorphin synthesis/release from leukocytes. However, the cellular mechanisms by which heme dampens β-endorphin production are inconclusive. The current hypothesis is that heme-dependent TLR4 activation and macrophage polarization to the M1 phenotype mediate this phenomenon. Our novel findings showed that PWH with CWP have elevated M1-specific macrophage chemokines (ENA-78, GRO-α, and IP-10) in plasma. In vitro, hemin-induced polarization of M0 and M2 macrophages to the M1 phenotype with low β-endorphins was mitigated by treating cells with the TLR4 inhibitor, TAK-242. Similarly, in vivo phenylhydrazine hydrochloride (PHZ), an inducer of hemolysis, injected into C57Bl/6 mice increased the M1/M2 cell ratio and reduced β-endorphin levels. However, treating these animals with the heme-scavenging protein hemopexin (Hx) or TAK-242 reduced the M1/M2 ratio and increased β-endorphins. Furthermore, Hx attenuated heme-induced mechanical, heat, and cold hypersensitivity, while TAK-242 abrogated hypersensitivity to mechanical and heat stimuli. Overall, these results suggest that heme-mediated TLR4 activation and M1 polarization of macrophages correlate with impaired endogenous opioid homeostasis and hypersensitivity in people with HIV.
Collapse
Affiliation(s)
- Tanima Chatterjee
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, PBMR 230, 901 19th Street South, Birmingham, AL 35205, USA; (T.C.); (L.B.U.); (T.L.L.); (J.X.M.J.)
| | - Itika Arora
- Division of Developmental Biology and the Reproductive Sciences Center, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA;
| | - Lilly B. Underwood
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, PBMR 230, 901 19th Street South, Birmingham, AL 35205, USA; (T.C.); (L.B.U.); (T.L.L.); (J.X.M.J.)
| | - Terry L. Lewis
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, PBMR 230, 901 19th Street South, Birmingham, AL 35205, USA; (T.C.); (L.B.U.); (T.L.L.); (J.X.M.J.)
| | - Juan Xavier Masjoan Juncos
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, PBMR 230, 901 19th Street South, Birmingham, AL 35205, USA; (T.C.); (L.B.U.); (T.L.L.); (J.X.M.J.)
| | - Sonya L. Heath
- Division of Infectious Disease, University of Alabama at Birmingham, Birmingham, AL 35205, USA;
| | - Burel R. Goodin
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO 63130, USA;
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, PBMR 230, 901 19th Street South, Birmingham, AL 35205, USA; (T.C.); (L.B.U.); (T.L.L.); (J.X.M.J.)
| |
Collapse
|
37
|
Voltarelli VA, Alves de Souza RW, Miyauchi K, Hauser CJ, Otterbein LE. Heme: The Lord of the Iron Ring. Antioxidants (Basel) 2023; 12:antiox12051074. [PMID: 37237940 DOI: 10.3390/antiox12051074] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Heme is an iron-protoporphyrin complex with an essential physiologic function for all cells, especially for those in which heme is a key prosthetic group of proteins such as hemoglobin, myoglobin, and cytochromes of the mitochondria. However, it is also known that heme can participate in pro-oxidant and pro-inflammatory responses, leading to cytotoxicity in various tissues and organs such as the kidney, brain, heart, liver, and in immune cells. Indeed, heme, released as a result of tissue damage, can stimulate local and remote inflammatory reactions. These can initiate innate immune responses that, if left uncontrolled, can compound primary injuries and promote organ failure. In contrast, a cadre of heme receptors are arrayed on the plasma membrane that is designed either for heme import into the cell, or for the purpose of activating specific signaling pathways. Thus, free heme can serve either as a deleterious molecule, or one that can traffic and initiate highly specific cellular responses that are teleologically important for survival. Herein, we review heme metabolism and signaling pathways, including heme synthesis, degradation, and scavenging. We will focus on trauma and inflammatory diseases, including traumatic brain injury, trauma-related sepsis, cancer, and cardiovascular diseases where current work suggests that heme may be most important.
Collapse
Affiliation(s)
- Vanessa Azevedo Voltarelli
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rodrigo W Alves de Souza
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kenji Miyauchi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Carl J Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Leo Edmond Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
38
|
Gotardo ÉMF, Brito PL, Gushiken LFS, Chweih H, Leonardo FC, Costa FF, Conran N. Molecular and cellular effects of in vivo chronic intravascular hemolysis and anti-inflammatory therapeutic approaches. Vascul Pharmacol 2023; 150:107176. [PMID: 37116732 DOI: 10.1016/j.vph.2023.107176] [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: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Intravascular hemolysis (IVH) occurs in numerous inherited and acquired disorders, including sickle cell disease (SCD), malaria and sepsis. These diseases display unique symptoms, but often share complications, such as vasomotor dysfunction and pulmonary hypertension. Consequently, in vivo models are needed to study the effects of continuous intravascular hemolytic processes, independently of the molecular or extrinsic alteration that leads to erythrocyte destruction. We gave twice-weekly low-dose phenylhydrazine (LDPHZ) to C57BL/6 J mice for 4 weeks, and measured parameters indicative of anemia, hemoglobin-clearance pathways, inflammation and iron turnover, comparing these to those of a murine model of SCD, which displays associated IVH. LDPHZ administration provoked discreet anemia in mice and significant reticulocytosis, in association with hemoglobin/heme-clearance pathway protein depletion. Mice subjected to chronic hemolysis displayed elevated leukocyte counts and plasma levels of interleukin (IL)-1β, TNF-α, IL-6, soluble ICAM-1, endothelin-1 and anti-inflammatory IL-10, closely emulating alterations indicative of systemic inflammatory and endothelial activation in SCD, and confirming chronic IVH in itself as a serious complication. Discreet accelerations in hepatic and splenic iron turnover also occurred in LDPHZ mice, without alterations in liver damage markers. Examining the effects of two therapies on hemolysis-induced inflammation, the administration of hydroxyurea (and to a lesser extent, l-glutamine) significantly abrogated hemolytic inflammation in mice, without apparent inhibition of hemolysis. In conclusion, the isolation of chronic IVH, a common disease mechanism, using this model, may allow the study of hemolysis-specific sequelae at the cellular and systemic level, and the investigation of candidate agents that could potentially counter hemolytic inflammation.
Collapse
Affiliation(s)
- Érica M F Gotardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | - Pâmela L Brito
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Lucas F S Gushiken
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Hanan Chweih
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Flavia C Leonardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Fernando F Costa
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Nicola Conran
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
39
|
Sesti-Costa R, Costa FF, Conran N. Role of Macrophages in Sickle Cell Disease Erythrophagocytosis and Erythropoiesis. Int J Mol Sci 2023; 24:ijms24076333. [PMID: 37047304 PMCID: PMC10094208 DOI: 10.3390/ijms24076333] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a β-globin gene point mutation that results in the production of sickle hemoglobin that polymerizes upon deoxygenation, causing the sickling of red blood cells (RBCs). RBC deformation initiates a sequence of events leading to multiple complications, such as hemolytic anemia, vaso-occlusion, chronic inflammation, and tissue damage. Macrophages participate in extravascular hemolysis by removing damaged RBCs, hence preventing the release of free hemoglobin and heme, and triggering inflammation. Upon erythrophagocytosis, macrophages metabolize RBC-derived hemoglobin, activating mechanisms responsible for recycling iron, which is then used for the generation of new RBCs to try to compensate for anemia. In the bone marrow, macrophages can create specialized niches, known as erythroblastic islands (EBIs), which regulate erythropoiesis. Anemia and inflammation present in SCD may trigger mechanisms of stress erythropoiesis, intensifying RBC generation by expanding the number of EBIs in the bone marrow and creating new ones in extramedullary sites. In the current review, we discuss the distinct mechanisms that could induce stress erythropoiesis in SCD, potentially shifting the macrophage phenotype to an inflammatory profile, and changing their supporting role necessary for the proliferation and differentiation of erythroid cells in the disease. The knowledge of the soluble factors, cell surface and intracellular molecules expressed by EBI macrophages that contribute to begin and end the RBC’s lifespan, as well as the understanding of their signaling pathways in SCD, may reveal potential targets to control the pathophysiology of the disease.
Collapse
|
40
|
De Simone G, Varricchio R, Ruberto TF, di Masi A, Ascenzi P. Heme Scavenging and Delivery: The Role of Human Serum Albumin. Biomolecules 2023; 13:biom13030575. [PMID: 36979511 PMCID: PMC10046553 DOI: 10.3390/biom13030575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Heme is the reactive center of several metal-based proteins that are involved in multiple biological processes. However, free heme, defined as the labile heme pool, has toxic properties that are derived from its hydrophobic nature and the Fe-atom. Therefore, the heme concentration must be tightly controlled to maintain cellular homeostasis and to avoid pathological conditions. Therefore, different systems have been developed to scavenge either Hb (i.e., haptoglobin (Hp)) or the free heme (i.e., high-density lipoproteins (HDL), low-density lipoproteins (LDL), hemopexin (Hx), and human serum albumin (HSA)). In the first seconds after heme appearance in the plasma, more than 80% of the heme binds to HDL and LDL, and only the remaining 20% binds to Hx and HSA. Then, HSA slowly removes most of the heme from HDL and LDL, and finally, heme transits to Hx, which releases it into hepatic parenchymal cells. The Hx:heme or HSA:heme complexes are internalized via endocytosis mediated by the CD91 and CD71 receptors, respectively. As heme constitutes a major iron source for pathogens, bacteria have evolved hemophores that can extract and uptake heme from host proteins, including HSA:heme. Here, the molecular mechanisms underlying heme scavenging and delivery from HSA are reviewed. Moreover, the relevance of HSA in disease states associated with increased heme plasma concentrations are discussed.
Collapse
Affiliation(s)
- Giovanna De Simone
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Romualdo Varricchio
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Tommaso Francesco Ruberto
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
- Centro Linceo Interdisciplinare Beniamino Segre, Accademia Nazionale dei Lincei, 00165 Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
- Accademia Nazionale dei Lincei, 00165 Roma, Italy
| |
Collapse
|
41
|
Natural polyphenol-based nanoparticles for the treatment of iron-overload disease. J Control Release 2023; 356:84-92. [PMID: 36813037 DOI: 10.1016/j.jconrel.2023.02.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Iron-overload diseases are characterized by a variety of symptoms resulting from excessive iron stores, oxidative stress and consequent end-organ damage. Deferoxamine (DFO) is an iron-chelator that can protect tissues from iron-induced damage. However, its application is limited due to its low stability and weak free radical scavenging ability. Herein, natural polyphenols have been employed to enhance the protective efficacy of DFO through the construction of supramolecular dynamic amphiphiles, which self-assemble into spherical nanoparticles with excellent scavenging capacity against both iron (III) and reactive oxygen species (ROS). This class of natural polyphenols-assisted nanoparticles was found to exhibit enhanced protective efficacy both in vitro in an iron-overload cell model and in vivo in an intracerebral hemorrhage model. This strategy of constructing natural polyphenols- assisted nanoparticles could benefit the treatment of iron-overload related diseases with excessive accumulation of toxic or harmful substances.
Collapse
|
42
|
Planchais C, Noe R, Gilbert M, Lecerf M, Kaveri SV, Lacroix-Desmazes S, Roumenina LT, Dimitrov JD. Oxidized hemoglobin triggers polyreactivity and autoreactivity of human IgG via transfer of heme. Commun Biol 2023; 6:168. [PMID: 36774392 PMCID: PMC9922299 DOI: 10.1038/s42003-023-04535-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/26/2023] [Indexed: 02/13/2023] Open
Abstract
Intravascular hemolysis occurs in diverse pathological conditions. Extracellular hemoglobin and heme have strong pro-oxidative and pro-inflammatory potentials that can contribute to the pathology of hemolytic diseases. However, many of the effects of extracellular hemoglobin and heme in hemolytic diseases are still not well understood. Here we demonstrate that oxidized hemoglobin (methemoglobin) can modify the antigen-binding characteristics of human immunoglobulins. Thus, incubation of polyclonal or some monoclonal human IgG in the presence of methemoglobin results in an appearance of binding reactivities towards distinct unrelated self-proteins, including the protein constituent of hemoglobin i.e., globin. We demonstrate that a transfer of heme from methemoglobin to IgG is indispensable for this acquisition of antibody polyreactivity. Our data also show that only oxidized form of hemoglobin have the capacity to induce polyreactivity of antibodies. Site-directed mutagenesis of a heme-sensitive human monoclonal IgG1 reveals details about the mechanism of methemoglobin-induced antigen-binding polyreactivity. Further here we assess the kinetics and thermodynamics of interaction of a heme-induced polyreactive human antibody with hemoglobin and myoglobin. Taken together presented data contribute to a better understanding of the functions of extracellular hemoglobin in the context of hemolytic diseases.
Collapse
Affiliation(s)
- Cyril Planchais
- Laboratory of Humoral Immunology, Institut Pasteur, Université Paris Cité, INSERM U1222, 75015 Paris, France
| | - Remi Noe
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Marie Gilbert
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maxime Lecerf
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Srini V. Kaveri
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Sébastien Lacroix-Desmazes
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Lubka T. Roumenina
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Jordan D. Dimitrov
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
43
|
Wiatr M, Hadzhieva M, Lecerf M, Noé R, Justesen S, Lacroix-Desmazes S, Dragon-Durey MA, Dimitrov JD. Hyperoxidized Species of Heme Have a Potent Capacity to Induce Autoreactivity of Human IgG Antibodies. Int J Mol Sci 2023; 24:ijms24043416. [PMID: 36834827 PMCID: PMC9960230 DOI: 10.3390/ijms24043416] [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: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The interaction of some human antibodies with heme results in posttranslational acquisition of binding to various self- and pathogen-derived antigens. The previous studies on this phenomenon were performed with oxidized heme (Fe3+). In the present study, we elucidated the effect of other pathologically relevant species of heme, i.e., species that were formed after contact of heme with oxidizing agents such as hydrogen peroxide, situations in which heme's iron could acquire higher oxidation states. Our data reveal that hyperoxidized species of heme have a superior capacity to heme (Fe3+) in triggering the autoreactivity of human IgG. Mechanistic studies demonstrated that oxidation status of iron was of critical importance for the heme's effect on antibodies. We also demonstrated that hyperoxidized heme species interacted at higher affinities with IgG and that this binding occurred through a different mechanism as compared to heme (Fe3+). Regardless of their profound functional impact on the antigen-binding properties of antibodies, hyperoxidized species of heme did not affect Fc-mediated functions of IgG, such as binding to the neonatal Fc receptor. The obtained data contribute to a better understanding of the pathophysiological mechanism of hemolytic diseases and of the origin of elevated antibody autoreactivity in patients with some hemolytic disorders.
Collapse
Affiliation(s)
- Marie Wiatr
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maya Hadzhieva
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Rémi Noé
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Sune Justesen
- Immunitrack Aps, Lersoe Park Alle 42, 2100 Copenhagen, Denmark
| | - Sébastien Lacroix-Desmazes
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Marie-Agnès Dragon-Durey
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Service d’Immunologie Biologique, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75610 Paris, France
| | - Jordan D. Dimitrov
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Correspondence: ; Tel.: +33-144-278206
| |
Collapse
|
44
|
Salgar S, Bolívar BE, Flanagan JM, Anum SJ, Bouchier-Hayes L. The NLRP3 inflammasome fires up heme-induced inflammation in hemolytic conditions. Transl Res 2023; 252:34-44. [PMID: 36041706 DOI: 10.1016/j.trsl.2022.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 01/14/2023]
Abstract
Overactive inflammatory responses are central to the pathophysiology of many hemolytic conditions including sickle cell disease. Excessive hemolysis leads to elevated serum levels of heme due to saturation of heme scavenging mechanisms. Extracellular heme has been shown to activate the NLRP3 inflammasome, leading to activation of caspase-1 and release of pro-inflammatory cytokines IL-1β and IL-18. Heme also activates the non-canonical inflammasome pathway, which may contribute to NLRP3 inflammasome formation and leads to pyroptosis, a type of inflammatory cell death. Some clinical studies indicate there is a benefit to blocking the NLRP3 inflammasome pathway in patients with sickle cell disease and other hemolytic conditions. However, a thorough understanding of the mechanisms of heme-induced inflammasome activation is needed to fully leverage this pathway for clinical benefit. This review will explore the mechanisms of heme-induced NLRP3 inflammasome activation and the role of this pathway in hemolytic conditions including sickle cell disease.
Collapse
Affiliation(s)
- Suruchi Salgar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Beatriz E Bolívar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jonathan M Flanagan
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Shaniqua J Anum
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
45
|
Marchesani S, Bertaina V, Marini O, Cossutta M, Di Mauro M, Rotulo GA, Palma P, Sabatini L, Petrone MI, Frati G, Monteleone G, Palumbo G, Ceglie G. Inflammatory status in pediatric sickle cell disease: Unravelling the role of immune cell subsets. Front Mol Biosci 2023; 9:1075686. [PMID: 36703915 PMCID: PMC9871358 DOI: 10.3389/fmolb.2022.1075686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction: The mutation of the beta-globin gene that causes sickle cell disease (SCD) results in pleiotropic effects, such as hemolysis and vaso-occlusive crisis that can induce inflammatory mechanisms with deleterious consequences on the organism. Moreover, SCD patients display an increased susceptibility to infections. Few studies are currently available that evaluate a wide immunological profile in a pediatric population. This study proposes an evaluation of the immune profile in subjects with SCD in a pediatric population through a detailed analysis by flow cytometry. Methods and Materials: Peripheral blood samples from 53 pediatric patients with SCD (mean age 9.8 years, interquartile range 9 years) were obtained and then analyzed by flow cytometry, in order to evaluate changes in the immune populations compared to 40 healthy donors (mean age 7.3 years, interquartile range 9.5 years). Results: Our data showed an increase in neutrophils (with a reduction in the CD62L + subpopulation) and monocytes (with a decrease in HLA-DRlow monocytes) with normal values of lymphocytes in SCD patients. In the lymphocyte subpopulations analysis we observed lower values of CD4+ T cells (with higher number of memory and central memory T lymphocytes) with increased frequency of CD8+ T cells (with a predominant naive pattern). Moreover, we observed higher values of CD39+ Tregs and lower HLA-DR+ and CD39- T cells with an increased Th17, Th1-17 and Th2 response. Conclusion: We observed immunological alterations typical of an inflammatory status (increase in activated neutrophils and monocytes) associated with a peculiar Treg pattern (probably linked to a body attempt to minimize inflammation intrinsic to SCD). Furthermore, we highlighted a T helper pathway associated with inflammation in line with other studies. Our data showed that immunological markers may have an important role in the understanding the pathophysiology of SCD and in optimizing targeted therapeutic strategies for each patient.
Collapse
Affiliation(s)
- Silvio Marchesani
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,*Correspondence: Silvio Marchesani,
| | - Valentina Bertaina
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Olivia Marini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Women’s and Children’s Health Department, Hematology-Oncology Clinic and Laboratory, University of Padova, Padova, Italy
| | - Matilde Cossutta
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Margherita Di Mauro
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Gioacchino Andrea Rotulo
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children Hospital, IRCCS, Rome, Italy,Department of Neuroscience, Rehabilitation Ophthalmology Genetics Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Paolo Palma
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,Clinical and Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Letizia Sabatini
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Maria Isabella Petrone
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Giacomo Frati
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giulia Monteleone
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Palumbo
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giulia Ceglie
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
46
|
Kour D, Ali M, Khajuria P, Sharma K, Ghosh P, Kaur S, Mahajan S, Ramajayan P, Bharate SS, Bhardwaj S, Sawant SD, Reddy DS, Kumar A. Flurbiprofen inhibits heme induced NLRP3 inflammasome in Berkeley sickle cell disease mice. Front Pharmacol 2023; 14:1123734. [PMID: 37180702 PMCID: PMC10171431 DOI: 10.3389/fphar.2023.1123734] [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: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Sickle cell disease (SCD) is accompanied by several complications, which emanate from the sickling of erythrocytes due to a point mutation in the β-globin chain of hemoglobin. Sickled erythrocytes are unable to move smoothly through small blood capillaries and therefore, cause vaso occlusion and severe pain. Apart from pain, continuous lysis of fragile sickled erythrocytes leads to the release of heme, which is a strong activator of the NLRP3 inflammasome, thus producing chronic inflammation in sickle cell disease. In this study, we identified flurbiprofen among other COX-2 inhibitors to be a potent inhibitor of heme-induced NLRP3 inflammasome. We found that apart from being a nociceptive agent, flurbiprofen exerts a strong anti-inflammatory effect by suppressing NF-κB signaling, which was evidenced by reduced levels of TNF-α and IL-6 in wild-type and sickle cell disease Berkeley mice models. Our data further demonstrated the protective effect of flurbiprofen on liver, lungs, and spleen in Berkeley mice. The current sickle cell disease pain management regime relies mainly on opiate drugs, which is accompanied by several side effects without modifying the sickle cell disease-related pathology. Considering the potent role of flurbiprofen in inhibiting NLRP3 inflammasome and other inflammatory cytokines in sickle cell disease, our data suggests that it can be explored further for better sickle cell disease pain management along with the possibility of disease modification.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Mehboob Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Palash Ghosh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sukhleen Kaur
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Surbhi Mahajan
- Department of Pathology, Government Medical College, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Sonali S. Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai, India
| | - Subhash Bhardwaj
- Department of Pathology, Government Medical College, Jammu, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - D. Srinivasa Reddy
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| |
Collapse
|
47
|
Xiao L, Wang M, Shi Y, Xu Y, Gao Y, Zhang W, Wu Y, Deng H, Pan W, Wang W, Sun H. Secondary White Matter Injury Mediated by Neuroinflammation after Intracerebral Hemorrhage and Promising Therapeutic Strategies of Targeting the NLRP3 Inflammasome. Curr Neuropharmacol 2023; 21:669-686. [PMID: 36043798 PMCID: PMC10207923 DOI: 10.2174/1570159x20666220830115018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a neurological disease with high mortality and disability. Recent studies showed that white matter injury (WMI) plays an important role in motor dysfunction after ICH. WMI includes WMI proximal to the lesion and WMI distal to the lesion, such as corticospinal tract injury located at the cervical enlargement of the spinal cord after ICH. Previous studies have tended to focus only on gray matter (GM) injury after ICH, and fewer studies have paid attention to WMI, which may be one of the reasons for the poor outcome of previous drug treatments. Microglia and astrocyte-mediated neuroinflammation are significant mechanisms responsible for secondary WMI following ICH. The NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome activation, has been shown to exacerbate neuroinflammation and brain injury after ICH. Moreover, NLRP3 inflammasome is activated in microglia and astrocytes and exerts a vital role in microglia and astrocytes-mediated neuroinflammation. We speculate that NLRP3 inflammasome activation is closely related to the polarization of microglia and astrocytes and that NLRP3 inflammasome activation may exacerbate WMI by polarizing microglia and astrocytes to the pro-inflammatory phenotype after ICH, while NLRP3 inflammasome inhibition may attenuate WMI by polarizing microglia and astrocytes to the anti-inflammatory phenotype following ICH. Therefore, NLRP3 inflammasome may act as leveraged regulatory fulcrums for microglia and astrocytes polarization to modulate WMI and WM repair after ICH. This review summarized the possible mechanisms by which neuroinflammation mediated by NLRP3 inflammasome exacerbates secondary WMI after ICH and discussed the potential therapeutic targets.
Collapse
Affiliation(s)
- Linglong Xiao
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Mengqi Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yifeng Shi
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yangyang Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yuan Gao
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yang Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Hao Deng
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Pan
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Haitao Sun
- Department of Laboratory Medicine, Clinical Biobank Center, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
48
|
Nath KA, Singh RD, Croatt AJ, Adams CM. Heme Proteins and Kidney Injury: Beyond Rhabdomyolysis. KIDNEY360 2022; 3:1969-1979. [PMID: 36514409 PMCID: PMC9717624 DOI: 10.34067/kid.0005442022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
49
|
Del Giudice E, Sota J, Orlando F, Picciano L, Cimaz R, Cantarini L, Mauro A. Off-label use of canakinumab in pediatric rheumatology and rare diseases. Front Med (Lausanne) 2022; 9:998281. [PMID: 36330067 PMCID: PMC9622922 DOI: 10.3389/fmed.2022.998281] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Since the first success of interleukin-1 blockade in cryopyrin-associated periodic syndrome, the use of interleukin-1 inhibitors has expanded to other disorders, including off-label indications. In particular, canakinumab has been employed in an off-label fashion in several diseases such as rare monogenic autoinflammatory diseases and multifactorial autoinflammatory diseases, disclosing an excellent efficacy and good safety profile in pediatric patients unresponsive to standards of care. In addition, hyperferritinemic syndromes and complex disorders, as well as Kawasaki disease, uveitis, and other pediatric rare disorders, represent additional areas where canakinumab efficacy is worth exploring. Altogether, the results summarized below are of paramount importance in pediatric patients where a considerable proportion of treatments are prescribed off-label. This review focuses on the off-label use of canakinumab in pediatric patients affected by systemic immune-mediated diseases.
Collapse
Affiliation(s)
- Emanuela Del Giudice
- Pediatric and Neonatology Unit, Maternal and Child Department, Sapienza University of Rome, Polo Pontino, Latina, Italy
| | - Jurgen Sota
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy
| | - Francesca Orlando
- Pediatric Rheumatology Unit, Department of General and Emergency Pediatrics, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Ludovica Picciano
- Pediatric Emergency and Short Stay Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Rolando Cimaz
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy
| | - Angela Mauro
- Pediatric Rheumatology Unit, Department of Childhood and Developmental Medicine, Fatebenefratelli-Sacco Hospital, Milan, Italy
- *Correspondence: Angela Mauro
| |
Collapse
|
50
|
A Model Peptide Reveals Insights into the Interaction of Human Hemopexin with Heme. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10441-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AbstractUnder hemolytic conditions, toxic heme is scavenged by hemopexin. Recently, the heme-binding properties of hemopexin have been reassessed, which revealed a KD of ~ 0.32 nM as well as a stoichiometry of one to two heme molecules binding to hemopexin. A 66mer hemopexin-derived peptide that spans over three heme-binding motifs was used to verify the earlier suggested heme-recruiting mechanism. Herein, we employed spectroscopic and computational methods to substantiate the hypothesis of more than one heme molecule binding to hemopexin and to analyze the heme-binding mode. Both, hemopexin and the 66mer peptide, were found to bind heme in mixed penta- and hexacoordinated states, which strongly indicates that heme binding follows distinct criteria and increases rigidity of the peptide-heme complex. Additional in silico molecular dynamics simulations support these experimental findings and, thus, contribute to our understanding of the molecular basis of the heme-hemopexin interaction. This analysis provides further details for consideration of hemopexin in biomedical applications.
Collapse
|