1
|
Heilberg IP, Carvalho AB, Denburg MR. Between a Rock and a Short Place-The Impact of Nephrolithiasis on Skeletal Growth and Development Across the Lifespan. Curr Osteoporos Rep 2024; 22:576-589. [PMID: 39356465 DOI: 10.1007/s11914-024-00888-w] [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] [Accepted: 09/10/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW The impact of nephrolithiasis on skeletal growth and bone health across the life span of kidney stone formers is reviewed. MAIN FINDINGS Bone disease is an early event among kidney stone formers (SF), with distinct phenotypes according to each age, sex, menopausal status, dietary, hormonal and genetic factors. Nephrolithiasis-associated bone disorder is characterized by reduced bone mineral density (BMD) and histologically discloses low bone formation, high bone resorption and abnormal mineralization. Although hypercalciuria has been presumed to be pathogenic for bone loss in SF, the association of BMD with urinary calcium is not uniform in all studies. Hypocitraturia, metabolic disturbances, cytokines and receptors, growth factors and acid-base status may all influence skeletal outcomes. The potential link of bone disease with vascular calcification and cardiovascular disease among SF is discussed. The unique vulnerability of the younger skeleton to the effects of nephrolithiasis on attainment of peak bone mass and strength is highlighted and the association of bone loss with kidney stone formation early in life indicate the opportunity for intervention to reduce the risk of future bone fractures.
Collapse
Affiliation(s)
- Ita Pfeferman Heilberg
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, Rua Botucatu 740 - Vila Clementino, São Paulo, 04023-900, Brazil.
| | - Aluizio Barbosa Carvalho
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, Rua Botucatu 740 - Vila Clementino, São Paulo, 04023-900, Brazil
| | - Michelle R Denburg
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
- Division of Pediatric Nephrology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
2
|
Arima M, Ito K, Abe T, Oguma T, Asano K, Mukherjee M, Ueki S. Eosinophilic mucus diseases. Allergol Int 2024; 73:362-374. [PMID: 38594175 DOI: 10.1016/j.alit.2024.03.002] [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: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Eosinophilic inflammation is primarily characterized by type 2 immune responses against parasitic organisms. In the contemporary human being especially in developed countries, eosinophilic inflammation is strongly associated with allergic/sterile inflammation, and constitutes an undesired immune reaction. This situation is in stark contrast to neutrophilic inflammation, which is indispensable for the host defense against bacterial infections. Among eosinophilic inflammatory disorders, massive accumulation of eosinophils within mucus is observed in certain cases, and is often linked to the distinctive clinical finding of mucus with high viscosity. Eosinophilic mucus is found in a variety of diseases, including chronic allergic keratoconjunctivitis, chronic rhinosinusitis encompassing allergic fungal sinusitis, eosinophilic otitis media, eosinophilic sialodochitis, allergic bronchopulmonary aspergillosis/mycosis, eosinophilic plastic bronchitis, and eosinophilic asthma. In these pathological conditions, chronic inflammation and tissue remodeling coupled with irreversible organ damage due to persistent adhesion of toxic substances and luminal obstruction may impose a significant burden on the body. Eosinophils aggregate in the hyperconcentrated mucus together with cell-derived crystals, macromolecules, and polymers, thereby affecting the biophysical properties of the mucus. This review focuses on the clinically significant challenges of mucus and discusses the consequences of activated eosinophils on the mucosal surface that impact mucus and persistent inflammation.
Collapse
Affiliation(s)
- Misaki Arima
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Keisuke Ito
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomoe Abe
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Tsuyoshi Oguma
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Manali Mukherjee
- Department of Medicine, McMaster University & St Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Shigeharu Ueki
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan.
| |
Collapse
|
3
|
Su Y, Li S, Li X, Zhou JY, Chauhan VP, Li M, Su YH, Liu CM, Ren YF, Yin W, Rimer JD, Cai T. Tartronic Acid as a Potential Inhibitor of Pathological Calcium Oxalate Crystallization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400642. [PMID: 38647258 DOI: 10.1002/advs.202400642] [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: 01/17/2024] [Revised: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Kidney stones are a pervasive disease with notoriously high recurrence rates that require more effective treatment strategies. Herein, tartronic acid is introduced as an efficient inhibitor of calcium oxalate monohydrate (COM) crystallization, which is the most prevalent constituent of human kidney stones. A combination of in situ experimental techniques and simulations are employed to compare the inhibitory effects of tartronic acid with those of its molecular analogs. Tartronic acid exhibits an affinity for binding to rapidly growing apical surfaces of COM crystals, thus setting it apart from other inhibitors such as citric acid, the current preventative treatment for kidney stones. Bulk crystallization and in situ atomic force microscopy (AFM) measurements confirm the mechanism by which tartronic acid interacts with COM crystal surfaces and inhibits growth. These findings are consistent with in vivo studies that reveal the efficacy of tartronic acid is similar to that of citric acid in mouse models of hyperoxaluria regarding their inhibitory effect on stone formation and alleviating stone-related physical harm. In summary, these findings highlight the potential of tartronic acid as a promising alternative to citric acid for the management of calcium oxalate nephropathies, offering a new option for clinical intervention in cases of kidney stones.
Collapse
Affiliation(s)
- Yuan Su
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Si Li
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204, USA
| | - Xin Li
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Science, Nanjing University, Nanjing, 210036, China
| | - Jing-Ying Zhou
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Vraj P Chauhan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204, USA
| | - Meng Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Ya-Hui Su
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Chun-Mei Liu
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi-Fei Ren
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Wu Yin
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Science, Nanjing University, Nanjing, 210036, China
| | - Jeffrey D Rimer
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204, USA
| | - Ting Cai
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
4
|
Woulfe J, Munoz DG, Gray DA, Jinnah HA, Ivanova A. Inosine monophosphate dehydrogenase intranuclear inclusions are markers of aging and neuronal stress in the human substantia nigra. Neurobiol Aging 2024; 134:43-56. [PMID: 37992544 DOI: 10.1016/j.neurobiolaging.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
We explored mechanisms involved in the age-dependent degeneration of human substantia nigra (SN) dopamine (DA) neurons. Owing to its important metabolic functions in post-mitotic neurons, we investigated the developmental and age-associated changes in the purine biosynthetic enzyme inosine monophosphate dehydrogenase (IMPDH). Tissue microarrays prepared from post-mortem samples of SN from 85 neurologically intact participants humans spanning the age spectrum were immunostained for IMPDH combined with other proteins. SN DA neurons contained two types of IMPDH structures: cytoplasmic IMPDH filaments and intranuclear IMPDH inclusions. The former were not age-restricted and may represent functional units involved in sustaining purine nucleotide supply in these highly metabolically active cells. The latter showed age-associated changes, including crystallization, features reminiscent of pathological inclusion bodies, and spatial associations with Marinesco bodies; structures previously associated with SN neuron dysfunction and death. We postulate dichotomous roles for these two subcellularly distinct IMPDH structures and propose a nucleus-based model for a novel mechanism of SN senescence that is independent of previously known neurodegeneration-associated proteins.
Collapse
Affiliation(s)
- John Woulfe
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - David G Munoz
- Li Ka Shing Knowledge Institute & Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, St. Michael's Hospital, Unity Health, University of Toronto, Toronto, Ontario, Canada
| | - Douglas A Gray
- Center for Cancer Therapeutics, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Hyder A Jinnah
- Departments of Neurology, Human Genetics & Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alyona Ivanova
- The Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children and Neurosurgery Research Department, St. Michael's Hospital, Toronto Unity Health, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Nicoletti T, Bink A, Helmchen B, Briel N, Frontzek K, Vlad B, Gaspert A, Boudriot E, Jung HH, Reuss AM, Weller M, Hortobágyi T. Neurologic involvement in cystinosis: Focus on brain lesions and new evidence of four-repeat (4R-) Tau immunoreactivity. J Neurol Sci 2024; 456:122841. [PMID: 38101161 DOI: 10.1016/j.jns.2023.122841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/10/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Nephropathic cystinosis is a rare autosomal recessive storage disorder caused by CTNS gene mutations, leading to autophagy-lysosomal pathway impairment and cystine crystals accumulation. Neurologic involvement is highly variable and includes both neurodevelopmental and neurodegenerative disturbances, as well as focal neurologic deficits. By presenting longitudinal data of a 28-year-old patient with a large infratentorial lesion, we summarized the pathology, clinical and imaging features of neurological involvement in cystinosis patients. Brain damage in form of cystinosis-related cerebral lesions occurs in advanced disease phases and is characterized by the accumulation of cystine crystals, subsequent inflammation with vasculitis-like features, necrosis, and calcification. Epilepsy is a frequent comorbidity in affected individuals. Steroids might play a role in the symptomatic treatment of "stroke-like" episodes due to edematous-inflammatory lesions, but probably do not change the overall prognosis. Lifelong compliance to depleting therapy with cysteamine still represents the main therapeutic option. However, consequences of CTNS gene defects are not restricted to cystine accumulation. New evidence of four-repeat (4R-) Tau immunoreactivity suggests concurrent progressive neurodegeneration in cystinosis patients, highlighting the need of innovative therapeutic strategies, and shedding light on the crosstalk between proteinopathies and autophagy-lysosomal system defects. Eventually, emerging easily accessible biomarkers such as serum neurofilament light chains (NfL) might detect subclinical neurologic involvement in cystinosis patients.
Collapse
Affiliation(s)
- Tommaso Nicoletti
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland.
| | - Andrea Bink
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland; Department of Neuroradiology, University Hospital Zurich, Switzerland
| | - Birgit Helmchen
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Nils Briel
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland; Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Karl Frontzek
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland; Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Benjamin Vlad
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Ariana Gaspert
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Elisabeth Boudriot
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Hans Heinrich Jung
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Li D, Yuan S, Deng Y, Wang X, Wu S, Chen X, Li Y, Ouyang J, Lin D, Quan H, Fu X, Li C, Mao W. The dysregulation of immune cells induced by uric acid: mechanisms of inflammation associated with hyperuricemia and its complications. Front Immunol 2023; 14:1282890. [PMID: 38053999 PMCID: PMC10694226 DOI: 10.3389/fimmu.2023.1282890] [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: 08/29/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Changes in lifestyle induce an increase in patients with hyperuricemia (HUA), leading to gout, gouty arthritis, renal damage, and cardiovascular injury. There is a strong inflammatory response in the process of HUA, while dysregulation of immune cells, including monocytes, macrophages, and T cells, plays a crucial role in the inflammatory response. Recent studies have indicated that urate has a direct impact on immune cell populations, changes in cytokine expression, modifications in chemotaxis and differentiation, and the provocation of immune cells by intrinsic cells to cause the aforementioned conditions. Here we conducted a detailed review of the relationship among uric acid, immune response, and inflammatory status in hyperuricemia and its complications, providing new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Delun Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Siyu Yuan
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiyao Deng
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Xiaowan Wang
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Shouhai Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Xuesheng Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Yimeng Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Jianting Ouyang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Danyao Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Haohao Quan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Xinwen Fu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Chuang Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Wei Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| |
Collapse
|
7
|
Chen SY, Li YP, You YP, Zhang HR, Shi ZJ, Liang QQ, Yuan T, Xu R, Xu LH, Zha QB, Ou-Yang DY, He XH. Theaflavin mitigates acute gouty peritonitis and septic organ injury in mice by suppressing NLRP3 inflammasome assembly. Acta Pharmacol Sin 2023; 44:2019-2036. [PMID: 37221235 PMCID: PMC10545837 DOI: 10.1038/s41401-023-01105-7] [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: 11/30/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
Activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome plays important role in defending against infections, but its aberrant activation is causally linked to many inflammatory diseases, thus being a therapeutic target for these diseases. Theaflavin, one major ingredient of black tea, exhibits potent anti-inflammatory and anti-oxidative activities. In this study, we investigated the therapeutic effects of theaflavin against NLRP3 inflammasome activation in macrophages in vitro and in animal models of related diseases. We showed that theaflavin (50, 100, 200 μM) dose-dependently inhibited NLRP3 inflammasome activation in LPS-primed macrophages stimulated with ATP, nigericin or monosodium urate crystals (MSU), evidenced by reduced release of caspase-1p10 and mature interleukin-1β (IL-1β). Theaflavin treatment also inhibited pyroptosis as shown by decreased generation of N-terminal fragment of gasdermin D (GSDMD-NT) and propidium iodide incorporation. Consistent with these, theaflavin treatment suppressed ASC speck formation and oligomerization in macrophages stimulated with ATP or nigericin, suggesting reduced inflammasome assembly. We revealed that theaflavin-induced inhibition on NLRP3 inflammasome assembly and pyroptosis resulted from ameliorated mitochondrial dysfunction and reduced mitochondrial ROS production, thereby suppressing interaction between NLRP3 and NEK7 downstream of ROS. Moreover, we showed that oral administration of theaflavin significantly attenuated MSU-induced mouse peritonitis and improved the survival of mice with bacterial sepsis. Consistently, theaflavin administration significantly reduced serum levels of inflammatory cytokines including IL-1β and attenuated liver inflammation and renal injury of mice with sepsis, concomitant with reduced generation of caspase-1p10 and GSDMD-NT in the liver and kidney. Together, we demonstrate that theaflavin suppresses NLRP3 inflammasome activation and pyroptosis by protecting mitochondrial function, thus mitigating acute gouty peritonitis and bacterial sepsis in mice, highlighting a potential application in treating NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Si-Yuan Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ya-Ping Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yi-Ping You
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Rui Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zi-Jian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qi-Qi Liang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Tao Yuan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Rong Xu
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Li-Hui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| | - Dong-Yun Ou-Yang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| |
Collapse
|
8
|
Gelardi M, Giancaspro R, Duda L, Quaranta VN, Pizzulli C, Maiorano E, Di Canio FM, Ruzza A, Iannuzzi L, Quaranta NAA, Parisi F, Cassano M, Marzullo A. Eosinophil-mast cell pattern of intraepithelial infiltration as a marker of severity in CRSwNP. Sci Rep 2023; 13:12101. [PMID: 37495667 PMCID: PMC10372103 DOI: 10.1038/s41598-023-39149-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is defined as a Type 2 eosinophilic disease, while CRSsNP is considered a Type 1 neutrophilic disease. Since neutrophils are also activated in eosinophilic CRSwNP, the eosinophil-neutrophil dualism has been revaluated. Among the inflammatory cells infiltrating sinus-nasal tissues, the role of mast cells (MCs) is not already recognized, although Clinical-Cytological Grading, which defines the severity of CRSwNP, attributes to mixed eosinophil-MC forms of CRSwNP a greater risk of recurrence. We aimed to examine nasal polyps from both a cytological and histopathological point of view, to evaluate the presence and localization of MCs. Cytological and histological examination of 39 samples of nasal polyps were performed. Immunohistochemistry was used to evaluate the presence of Tryptase + CD117 + MCs, which were counted both in the epithelial layer and in the lamina propria. A statistically significant correlation was found between intraepithelial MCs and CRSwNP severity (p < 0.001) and between the total eosinophil count and the total mast cell count (p < 0.001). Cytological examination and immunohistochemistry were comparable in detecting the presence of intraepithelial MCs (p = 0.002). The histological cut-off of 6 intraepithelial MCs was identified to detect severe CRSwNP (p < 0.001). MCs have been shown to be located in the lamina propria of almost all eosinophilic nasal polyps without significantly affecting their severity. Intraepithelial MCs are associated with greater severity of CRSwNP. Histopathological criteria of the eosinophil-MC form of CRSwNP in addition to the eosinophilic one, should be defined to guarantee patients effective and tailored treatments.
Collapse
Affiliation(s)
- Matteo Gelardi
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto 1, 71122, Foggia, Italy
| | - Rossana Giancaspro
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto 1, 71122, Foggia, Italy.
| | - Loren Duda
- Unit of Pathology, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Vitaliano Nicola Quaranta
- Section of Respiratory Disease, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Cristina Pizzulli
- Unit of Pathology, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Eugenio Maiorano
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Filomena Milena Di Canio
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto 1, 71122, Foggia, Italy
| | - Annamaria Ruzza
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto 1, 71122, Foggia, Italy
| | - Lucia Iannuzzi
- Otolaryngology Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, University of Bari Aldo Moro, Bari, Italy
| | - Nicola Antonio Adolfo Quaranta
- Otolaryngology Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, University of Bari Aldo Moro, Bari, Italy
| | | | - Michele Cassano
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto 1, 71122, Foggia, Italy
| | - Andrea Marzullo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari "Aldo Moro", 70124, Bari, Italy
| |
Collapse
|
9
|
Zheng B, He Y, Yin S, Zhu X, Zhao Q, Yang H, Wang Z, Zhu R, Cheng L. Unresolved Excess Accumulation of Myelin-Derived Cholesterol Contributes to Scar Formation after Spinal Cord Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0135. [PMID: 37223476 PMCID: PMC10202378 DOI: 10.34133/research.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/25/2023]
Abstract
Spinal cord injury triggers complex pathological cascades, resulting in destructive tissue damage and incomplete tissue repair. Scar formation is generally considered a barrier for regeneration in the central nervous system. However, the intrinsic mechanism of scar formation after spinal cord injury has not been fully elucidated. Here, we report that excess cholesterol accumulates in phagocytes and is inefficiently removed from spinal cord lesions in young adult mice. Interestingly, we observed that excessive cholesterol also accumulates in injured peripheral nerves but is subsequently removed by reverse cholesterol transport. Meanwhile, preventing reverse cholesterol transport leads to macrophage accumulation and fibrosis in injured peripheral nerves. Furthermore, the neonatal mouse spinal cord lesions are devoid of myelin-derived lipids and can heal without excess cholesterol accumulation. We found that transplantation of myelin into neonatal lesions disrupts healing with excessive cholesterol accumulation, persistent macrophage activation, and fibrosis. Myelin internalization suppresses macrophage apoptosis mediated by CD5L expression, indicating that myelin-derived cholesterol plays a critical role in impaired wound healing. Taken together, our data suggest that the central nervous system lacks an efficient approach for cholesterol clearance, resulting in excessive accumulation of myelin-derived cholesterol, thereby inducing scar formation after injury.
Collapse
Affiliation(s)
- Bolin Zheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Yijing He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Shuai Yin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Xu Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Qing Zhao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Clinical Center for Brain and Spinal Cord Research,
Tongji University, Shanghai 200092, China
| |
Collapse
|
10
|
Gelardi M, Giancaspro R, Cassano M. Charcot-Leyden crystals: An ancient but never so current discovery. Am J Otolaryngol 2023; 44:103844. [PMID: 36948077 DOI: 10.1016/j.amjoto.2023.103844] [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: 12/15/2022] [Revised: 02/27/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
From the first description of Charcot-Leyden crystals (CLCs) to the present, many steps have been taken to understand the mechanisms underlying their formation. In particular, to date not only eosinophils but also mast cells are known to be responsible for the production of CLCs, which represent the crystallized form of Galectin-10. Due to their characteristics, CLCs typically induce a crystallopathy and are responsible for an exacerbation of inflammation. Nasal cytology (NC) has allowed to better understand the correlation between the severity of several rhinopaties and the presence of CLCs in NC samples, which is strictly correlated with an eosinophiles and mast cells infiltration. As a matter of fact, rhinopaties with a mixed eosinophilic-mast cell inflammatory infiltrate, characterized by the presence of abundant CLCs, show a worse prognosis and a higher risk of relapse. This could have important therapeutic implications, since the treatments available today could be exploited to target both eosinophils and mast cells, to reduce the damage induced by CLCs.
Collapse
Affiliation(s)
- M Gelardi
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - R Giancaspro
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - M Cassano
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW This short review is intended to highlight the potential role of inflammation as a key pathological driver, rather than a mere consequence, of nephrolithiasis. Although there is clearly a strong likelihood that the relationship is bidirectional, and that kidney stone-triggered inflammation can establish a vicious cycle of tissue injury and stone formation. RECENT FINDINGS These consist of data from both recent preclinical and clinical studies demonstrating the importance of inflammation in models of stone disease and in kidney tissue from patients with nephrolithiasis, and as a potential driver of disease recurrence and a suitable treatment target. In particular, the role of immune cells and their relationship to the NLRP3 inflammasome is becoming clearer, as well as the potential contribution to tissue injury and stone formation of the pro-inflammatory cytokines interleukin-1β and interleukin-18. SUMMARY This concept is not new and raises the possibility that targeting inflammation directly may prove to be a novel and suitable means of treatment for at least some types of kidney stone, and in certain clinical settings, both acutely and as prevention, especially in those patients experiencing recurrent stone episodes and/or who have a well defined metabolic cause such as uric acid or calcium oxalate stones.
Collapse
Affiliation(s)
- Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples
| | - Pietro Manuel Ferraro
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London (UCL), London, UK
| |
Collapse
|
12
|
Fadeeva IS, Teterina AY, Minaychev VV, Senotov AS, Smirnov IV, Fadeev RS, Smirnova PV, Menukhov VO, Lomovskaya YV, Akatov VS, Barinov SM, Komlev VS. Biomimetic Remineralized Three-Dimensional Collagen Bone Matrices with an Enhanced Osteostimulating Effect. Biomimetics (Basel) 2023; 8:biomimetics8010091. [PMID: 36975321 PMCID: PMC10046016 DOI: 10.3390/biomimetics8010091] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Bone grafts with a high potential for osseointegration, capable of providing a complete and effective regeneration of bone tissue, remain an urgent and unresolved issue. The presented work proposes an approach to develop composite biomimetic bone material for reconstructive surgery by deposition (remineralization) on the surface of high-purity, demineralized bone collagen matrix calcium phosphate layers. Histological and elemental analysis have shown reproduction of the bone tissue matrix architectonics, and a high-purity degree of the obtained collagen scaffolds; the cell culture and confocal microscopy have demonstrated a high biocompatibility of the materials obtained. Adsorption spectroscopy, scanning electron microscopy, microcomputed tomography (microCT) and infrared spectroscopy, and X-ray diffraction have proven the efficiency of the deposition of calcium phosphates on the surface of bone collagen scaffolds. Cell culture and confocal microscopy methods have shown high biocompatibility of both demineralized and remineralized bone matrices. In the model of heterotopic implantation in rats, at the term of seven weeks, an intensive intratrabecular infiltration of calcium phosphate precipitates, and a pronounced synthetic activity of osteoblast remodeling and rebuilding implanted materials, were revealed in remineralized bone collagen matrices in contrast to demineralized ones. Thus, remineralization of highly purified demineralized bone matrices significantly enhanced their osteostimulating ability. The data obtained are of interest for the creation of new highly effective osteoplastic materials for bone tissue regeneration and augmentation.
Collapse
Affiliation(s)
- Irina S. Fadeeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| | - Anastasia Yu. Teterina
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| | - Vladislav V. Minaychev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Anatoliy S. Senotov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Igor V. Smirnov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Roman S. Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Polina V. Smirnova
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Vladislav O. Menukhov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Yana V. Lomovskaya
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Vladimir S. Akatov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Sergey M. Barinov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Vladimir S. Komlev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| |
Collapse
|
13
|
Stamatelou K, Goldfarb DS. Epidemiology of Kidney Stones. Healthcare (Basel) 2023; 11:healthcare11030424. [PMID: 36766999 PMCID: PMC9914194 DOI: 10.3390/healthcare11030424] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
In the past two decades, major breakthroughs that improve our understanding of the pathophysiology and therapy of kidney stones (KS) have been lacking. The disease continues to be challenging for patients, physicians, and healthcare systems alike. In this context, epidemiological studies are striving to elucidate the worldwide changes in the patterns and the burden of the disease and identify modifiable risk factors that contribute to the development of kidney stones. Our expanding knowledge of the epidemiology of kidney stones is of paramount importance and largely upgrades the modern management of the disease. In this paper, we review the variables affecting prevalence and incidence, including age, gender, race, ethnicity, occupation, climate, geography, systemic diseases, diabetes, vascular disease, chronic kidney disease, and dietary risk factors relevant to kidney stones.
Collapse
Affiliation(s)
- Kyriaki Stamatelou
- “MESOGEIOS” Nephrology Center, Haidari and Nephros.eu Private Clinic, 11527 Athens, Greece
| | - David S. Goldfarb
- Nephrology Division, NYU Langone Health and NYU Grossman School of Medicine, NY Nephrology Section, NY Harbor VA Healthcare System, New York, NY 10016, USA
- Correspondence: ; Tel.: +1-212-686-7500 (ext. 3877); Fax: +1-212-951-6842
| |
Collapse
|
14
|
Abstract
Cell death, particularly that of tubule epithelial cells, contributes critically to the pathophysiology of kidney disease. A body of evidence accumulated over the past 15 years has ascribed a central pathophysiological role to a particular form of regulated necrosis, termed necroptosis, to acute tubular necrosis, nephron loss and maladaptive renal fibrogenesis. Unlike apoptosis, which is a non-immunogenic process, necroptosis results in the release of cellular contents and cytokines, which triggers an inflammatory response in neighbouring tissue. This necroinflammatory environment can lead to severe organ dysfunction and cause lasting tissue injury in the kidney. Despite evidence of a link between necroptosis and various kidney diseases, there are no available therapeutic options to target this process. Greater understanding of the molecular mechanisms, triggers and regulators of necroptosis in acute and chronic kidney diseases may identify shortcomings in current approaches to therapeutically target necroptosis regulators and lead to the development of innovative therapeutic approaches.
Collapse
|
15
|
Tomizawa H, Yamada Y, Arima M, Miyabe Y, Fukuchi M, Hikichi H, Melo RCN, Yamada T, Ueki S. Galectin-10 as a Potential Biomarker for Eosinophilic Diseases. Biomolecules 2022; 12:biom12101385. [PMID: 36291593 PMCID: PMC9599181 DOI: 10.3390/biom12101385] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Galectin-10 is a member of the lectin family and one of the most abundant cytoplasmic proteins in human eosinophils. Except for some myeloid leukemia cells, basophils, and minor T cell populations, galectin-10 is exclusively present in eosinophils in the human body. Galectin-10 forms Charcot–Leyden crystals, which are observed in various eosinophilic diseases. Accumulating studies have indicated that galectin-10 acts as a new biomarker for disease activity, diagnosis, and treatment effectiveness in asthma, eosinophilic esophagitis, rhinitis, sinusitis, atopic dermatitis, and eosinophilic granulomatosis with polyangiitis. The extracellular release of galectin-10 is not mediated through conventional secretory processes (piecemeal degranulation or exocytosis), but rather by extracellular trap cell death (ETosis), which is an active cell death program. Eosinophils undergoing ETosis rapidly disintegrate their plasma membranes to release the majority of galectin-10. Therefore, elevated galectin-10 levels in serum and tissue suggest a high degree of eosinophil ETosis. To date, several studies have shown that galectin-10/Charcot–Leyden crystals are more than just markers for eosinophilic inflammation, but play functional roles in immunity. In this review, we focus on the close relationship between eosinophils and galectin-10, highlighting this protein as a potential new biomarker in eosinophilic diseases.
Collapse
Affiliation(s)
- Hiroki Tomizawa
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
- Department of Otorhinolaryngology, Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yoshiyuki Yamada
- Department of Pediatrics, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Misaki Arima
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yui Miyabe
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
- Department of Otorhinolaryngology, Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Mineyo Fukuchi
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Haruka Hikichi
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Rossana C. N. Melo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil
| | - Takechiyo Yamada
- Department of Otorhinolaryngology, Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Shigeharu Ueki
- Clinical Laboratory Medicine, Department of General Internal Medicine, Akita University Graduate School of Medicine, Akita 010-8543, Japan
- Correspondence: ; Tel./Fax: +81-18-884-6209
| |
Collapse
|
16
|
Targeting innate immunity-driven inflammation in CKD and cardiovascular disease. Nat Rev Nephrol 2022; 18:762-778. [PMID: 36064794 DOI: 10.1038/s41581-022-00621-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 11/08/2022]
Abstract
Mortality among patients with chronic kidney disease (CKD) is largely a consequence of cardiovascular disease (CVD) and is a particular concern given the increasing prevalence of CKD. Sterile inflammation triggered by activation of the innate immune system is an important driver of both CKD and associated CVD. Several endogenous mediators, including lipoproteins, crystals such as silica, urate and cholesterol crystals, or compounds released from dying cells interact with pattern recognition receptors expressed on a variety of different cell types, leading to the release of pro-inflammatory cytokines. Disturbed regulation of the haematopoietic system by damage-associated molecular patterns, or as a consequence of clonal haematopoiesis or trained innate immunity, also contributes to the development of inflammation. In observational and genetic association studies, inflammation is linked to the progression of CKD and cardiovascular events. In 2017, the CANTOS trial of canakinumab provided evidence that inhibiting inflammation driven by NLRP3-IL-1-IL-6-mediated signalling significantly reduced cardiovascular event rates in individuals with and without CKD. Other approaches to target innate immune pathways are now under investigation for their ability to reduce cardiovascular events and slow disease progression among patients with atherosclerosis and stage 3 and 4 CKD. This Review summarizes current understanding of the role of inflammation in the pathogenesis of CKD and its associated CVD, and how this knowledge may translate into novel therapeutics.
Collapse
|
17
|
Colboc H, Bettuzzi T, Badrignans M, Bazin D, Boury A, Letavernier E, Frochot V, Tang E, Moguelet P, Ortonne N, de Prost N, Ingen-Housz-Oro S, Daudon M. Relationship between calcinosis cutis in epidermal necrolysis and caspofungin, a physicochemical investigation. CR CHIM 2022. [DOI: 10.5802/crchim.202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
18
|
Bazin D, Daudon M, Frochot V, Haymann JP, Letavernier E. Foreword to microcrystalline pathologies: combining clinical activity and fundamental research at the nanoscale. CR CHIM 2022. [DOI: 10.5802/crchim.200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Bazin D, Lucas IT, Rouzière S, Elkaim E, Mocuta C, Réguer S, Reid DG, Mathurin J, Dazzi A, Deniset-Besseau A, Petay M, Frochot V, Haymann JP, Letavernier E, Verpont MC, Foy E, Bouderlique E, Colboc H, Daudon M. Profile of an “at cutting edge” pathology laboratory for pathological human deposits: from nanometer to in vivo scale analysis on large scale facilities. CR CHIM 2022. [DOI: 10.5802/crchim.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Bao D, Wang Y, Yu X, Zhao M. Acute oxalate nephropathy: A potential cause of acute kidney injury in diabetes mellitus—A case series from a single center. Front Med (Lausanne) 2022; 9:929880. [PMID: 36133577 PMCID: PMC9484473 DOI: 10.3389/fmed.2022.929880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAcute oxalate nephropathy (AON) is an uncommon condition that causes acute kidney injury (AKI), characterized by the massive deposition of calcium oxalate crystals in the renal parenchyma. In previous studies, urinary oxalate excretion has been found to be increased in patients with diabetes mellitus (DM). Here, we report a case series of diabetic patients with AKI with biopsy-proven AON, aiming to alert physicians to the potential of AON as a trigger of AKI in diabetic patients in clinical practice.Materials and methodsCases with pathological diagnosis of AON who presented with AKI clinically and had DM between January 2016 and December 2020 were retrospectively enrolled. Their clinical and pathological manifestations, treatment, and prognosis were collected.ResultsSix male patients with biopsy-proven AON out of a total of 5,883 native kidney biopsies were identified, aged 58.3 ± 9.1 years at the time of kidney biopsy. Only one patient who had received Roux-en-Y gastric bypass surgery took oxalate-rich food before the onset of the disease. None of them had clinical features of enteric malabsorption. Three patients were currently on renin-angiotensin system inhibitor treatment for hypertension, and 5 of them received non-steroidal anti-inflammatory drugs. Three patients presented with oliguria and 4 patients needed dialysis at the beginning with none requiring dialysis at discharge. Four patients received a course of corticosteroid treatment empirically. Among them, two patients had estimated glomerular filtration rate (eGFR) recovered to over 60 ml/min/1.73 m2, while the other two patients remained with kidney dysfunction at the last follow-up. In two patients without corticosteroid treatment, one patient fully recovered with eGFR over 90 ml/min/1.73 m2 and the other patient remained with kidney dysfunction at the last follow-up.ConclusionAON might be a rare but potentially trigger of AKI in patients with DM. A kidney biopsy could help physicians to make the correct diagnosis. The proper treatment to alleviate oxalate-induced injury needs to be further studied.
Collapse
Affiliation(s)
- Daorina Bao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, National Health and Family Planning Commission of the People’s Republic of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yu Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, National Health and Family Planning Commission of the People’s Republic of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- *Correspondence: Yu Wang,
| | - Xiaojuan Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, National Health and Family Planning Commission of the People’s Republic of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, National Health and Family Planning Commission of the People’s Republic of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Laboratory of Electron Microscopy, Pathological Centre, Peking University First Hospital, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
21
|
Srivastava A, Tomar B, Sharma P, Kumari S, Prakash S, Rath SK, Kulkarni OP, Gupta SK, Mulay SR. RIPK3-MLKL signaling activates mitochondrial CaMKII and drives intrarenal extracellular matrix production during CKD. Matrix Biol 2022; 112:72-89. [PMID: 35964866 DOI: 10.1016/j.matbio.2022.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/19/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022]
Abstract
Intrarenal extracellular matrix production is a prevalent feature of all forms of chronic kidney disease (CKD). The transforming growth factor-beta (TGFβ) is believed to be a major driver of extracellular matrix production. Nevertheless, anti-TGFβ therapies have consistently failed to reduce extracellular matrix production in CKD patients indicating the need for novel therapeutic strategies. We have previously shown that necroinflammation contributes to acute kidney injury. Here, we show that chronic/persistent necroinflammation drives intrarenal extracellular matrix production during CKD. We found that renal expression of receptor-interacting protein kinase-1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) increases with the expansion of intrarenal extracellular matrix production and declined kidney function in both humans and mice. Furthermore, we found that TGFβ exposure induces the translocation of RIPK3 and MLKL to mitochondria resulting in mitochondrial dysfunction and ROS production. Mitochondrial ROS activates the serine-threonine kinase calcium/calmodulin-dependent protein kinases-II (CaMKII) that increases phosphorylation of Smad2/3 and subsequent production of alpha-smooth muscle actin (αSMA), collagen (Col) 1α1, etc. in response to TGFβ during the intrarenal extracellular matrix production. Consistent with this, deficiency or knockdown of RIPK3 or MLKL as well as pharmacological inhibition of RIPK1, RIPK3, and CaMKII prevents the intrarenal extracellular matrix production in oxalate-induced CKD and unilateral ureteral obstruction (UUO). Together, RIPK1, RIPK3, MLKL, CaMKII, and Smad2/3 are molecular targets to inhibit intrarenal extracellular matrix production and preserve kidney function during CKD.
Collapse
Affiliation(s)
- Anjali Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Bhawna Tomar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, 500078, India
| | - Sunaina Kumari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shakti Prakash
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srikanta Kumar Rath
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, 500078, India
| | - Shashi Kumar Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shrikant R Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
22
|
Noda H, Yutani C, Zaima N, Komatsu S, Iwa N, Takahashi S, Takewa M, Ohara T, Kodama K. Detection of macrophages engulfing cholesterol crystals and docosahexaenoic acid from spontaneous ruptured aortic plaque. J Cardiol Cases 2022; 26:364-366. [DOI: 10.1016/j.jccase.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022] Open
|
23
|
Collins MT, Marcucci G, Anders HJ, Beltrami G, Cauley JA, Ebeling PR, Kumar R, Linglart A, Sangiorgi L, Towler DA, Weston R, Whyte MP, Brandi ML, Clarke B, Thakker RV. Skeletal and extraskeletal disorders of biomineralization. Nat Rev Endocrinol 2022; 18:473-489. [PMID: 35578027 DOI: 10.1038/s41574-022-00682-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 12/15/2022]
Abstract
The physiological process of biomineralization is complex and deviation from it leads to a variety of diseases. Progress in the past 10 years has enhanced understanding of the genetic, molecular and cellular pathophysiology underlying these disorders; sometimes, this knowledge has both facilitated restoration of health and clarified the very nature of biomineralization as it occurs in humans. In this Review, we consider the principal regulators of mineralization and crystallization, and how dysregulation of these processes can lead to human disease. The knowledge acquired to date and gaps still to be filled are highlighted. The disorders of mineralization discussed comprise a broad spectrum of conditions that encompass bone disorders associated with alterations of mineral quantity and quality, as well as disorders of extraskeletal mineralization (hyperphosphataemic familial tumoural calcinosis). Included are disorders of alkaline phosphatase (hypophosphatasia) and phosphate homeostasis (X-linked hypophosphataemic rickets, fluorosis, rickets and osteomalacia). Furthermore, crystallopathies are covered as well as arterial and renal calcification. This Review discusses the current knowledge of biomineralization derived from basic and clinical research and points to future studies that will lead to new therapeutic approaches for biomineralization disorders.
Collapse
Affiliation(s)
- Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA.
| | - Gemma Marcucci
- Bone Metabolic Diseases Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Hans-Joachim Anders
- Department of Medicine IV, Hospital of the University of Munich, Ludwig-Maximilians University, Munich, Germany
| | - Giovanni Beltrami
- Department Paediatric Orthopedic Oncology, Careggi and Meyer Children Hospital, Florence, Italy
| | - Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Rajiv Kumar
- Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Agnès Linglart
- APHP, Endocrinologie et diabète de l'enfant, Paris, France
| | - Luca Sangiorgi
- Medical Genetics and Skeletal Rare Diseases, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Dwight A Towler
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ria Weston
- Cardiovascular Research Group, Manchester Metropolitan University, Manchester, UK
| | - Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospitals for Children-St Louis, St Louis, MO, USA
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | - Bart Clarke
- Mayo Clinic Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Rochester, MN, USA
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Gout-associated monosodium urate crystal-induced necrosis is independent of NLRP3 activity but can be suppressed by combined inhibitors for multiple signaling pathways. Acta Pharmacol Sin 2022; 43:1324-1336. [PMID: 34376811 PMCID: PMC9061757 DOI: 10.1038/s41401-021-00749-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Monosodium urate (MSU) crystals, the etiological agent of gout, are formed in joints and periarticular tissues due to long-lasting hyperuricemia. Although MSU crystal-triggered NLRP3 inflammasome activation and interleukin 1β (IL-1β) release are known to have key roles in gouty arthritis, recent studies revealed that MSU crystal-induced necrosis also plays a critical role in this process. However, it remains unknown what forms of necrosis have been induced and whether combined cell death inhibitors can block such necrosis. Here, we showed that MSU crystal-induced necrosis in murine macrophages was not dependent on NLRP3 inflammasome activation, as neither genetic deletion nor pharmacological blockade of the NLRP3 pathway inhibited the necrosis. Although many cell death pathways (such as ferroptosis and pyroptosis) inhibitors or reactive oxygen species inhibitors did not have any suppressive effects, necroptosis pathway inhibitors GSK'872 (RIPK3 inhibitor), and GW806742X (MLKL inhibitor) dose-dependently inhibited MSU crystal-induced necrosis. Moreover, a triple combination of GSK'872, GW806742X, and IDN-6556 (pan-caspase inhibitor) displayed enhanced inhibition of the necrosis, which was further fortified by the addition of MCC950 (NLRP3 inhibitor), suggesting that multiple cell death pathways might have been triggered by MSU crystals. Baicalin, a previously identified inhibitor of NLRP3, inhibited MSU crystal-induced inflammasome activation and suppressed the necrosis in macrophages. Besides, baicalin gavage significantly ameliorated MSU crystal-induced peritonitis in mice. Altogether, our data indicate that MSU crystals induce NLRP3-independent necrosis, which can be inhibited by combined inhibitors for multiple signaling pathways, highlighting a new avenue for the treatment of gouty arthritis.
Collapse
|
25
|
Vignes S, Fourgeaud C, Michon-Pasturel U. Un syndrome des orteils bleus révélant un cancer de la prostate métastatique : une observation avec analyse de la littérature. Rev Med Interne 2022; 43:562-565. [DOI: 10.1016/j.revmed.2022.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/02/2022] [Accepted: 04/09/2022] [Indexed: 11/25/2022]
|
26
|
Testa A, Chamorey E, Lavainne F, Verger C. Outcomes of patients with Cholesterol crystal embolism treated by peritoneal dialysis: retrospective study from the RDPLF registry. BULLETIN DE LA DIALYSE À DOMICILE 2022. [DOI: 10.25796/bdd.v5i1.65303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cholesterol crystal embolisms disease (CED) is a systemic disease characterized by tissue ischemia and microinflammation related to occlusion of arterioles by microemboli from ulcerated atheromatous plaques. The kidney is one of the most affected organs, often requiring dialysis. Our study examines the outcomes of a cohort of patients with CED treated with peritoneal dialysis (PD). As anticoagulants can favor emboli, peritoneal dialysis may theoretically have an advantage.
From the database of the French Language Peritoneal Dialysis Registry (RDPLF), between January 1, 1995, and December 31, 2021, we selected patients treated with PD for >90 days and with an age >18 years. On the basis of the variables of patient autonomy, diabetes, BMI, modified Charlson index, age, sex, pre-PD treatment, causes of death, causes of transfer to hemodialysis, and peritonitis, three types of survival were estimated (patient, technical, and composite). After adjustment by propensity scores and taking into account competitive risks, 2 groups of patients were constituted on the basis of baseline nephropathy: emboli group vs. control group.
Patient survival and strict technical survival are not associated with the type of nephropathy (CED versus others). Composite technical survival (uncensored for deaths and transfers to hemodialysis) was only associated with cholesterol emboli nephropathy in the multivariate analysis adjusted for diabetes, autonomy, and age of the patient at the start of the dialysis treatment (p=0.011; 95% CI [0.736 [0.581-0.931]]).
Our study from the RDPLF database shows no difference in technical and patient survival in a cohort of patients with CED vs. a control group. It also confirms that PD may represent an adequate choice in this pathology.
Collapse
|
27
|
Gu Y, Shen Y, Chen W, He H, Ma Y, Mei X, Ju D, Liu H. Protective effects of interleukin-22 on oxalate-induced crystalline renal injury via alleviating mitochondrial damage and inflammatory response. Appl Microbiol Biotechnol 2022; 106:2637-2649. [PMID: 35294590 DOI: 10.1007/s00253-022-11876-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/06/2022] [Indexed: 11/30/2022]
Abstract
Oxalate-induced crystalline kidney injury is one of the most common types of crystalline nephropathy. Unfortunately, there is no effective treatment to reduce the deposition of calcium oxalate crystals and alleviate kidney damage. Thus, proactive therapeutic is urgently needed to alleviate the suffering it causes to patient. Here, we investigated whether IL-22 exerted nephroprotective effects to sodium oxalate-mediated kidney damage and its potential mechanism. Crystalline kidney injury models were developed in vitro and in vivo that was often observed in clinic. We provided evidence that IL-22 could effectively decrease the accumulation of ROS and mitochondrial damage in cell and animal models and reduce the death of TECs. Moreover, IL-22 decreased the expression of the NLRP3 inflammasome and mature IL-1β in renal tissue induced by sodium oxalate. Further studies confirmed that IL-22 could play an anti-inflammatory role by reducing the levels of cytokines such as IL-1β, IL-18, and TNF-α in serum. In conclusion, our study confirmed that IL-22 has protective effects on sodium oxalate-induced crystalline kidney injury by reducing the production of ROS, protecting mitochondrial membrane potential, and inhibiting the inflammatory response. Therefore, IL-22 may play a potential preventive role in sodium oxalate-induced acute renal injury. KEY POINTS: • IL-22 could reduce sodium oxalate-mediated cytotoxicity and ameliorate renal injury. • IL-22 could alleviate oxidative stress and mitochondrial dysfunction induced by sodium oxalate. • IL-22 could inhibit inflammatory response of renal injury caused by sodium oxalate.
Collapse
Affiliation(s)
- Yuting Gu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Yilan Shen
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Wei Chen
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai, 201203, China
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China
| | - Haidong He
- Department of Nephrology, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, China
| | - Yulei Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Xiaobin Mei
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Dianwen Ju
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai, 201203, China.
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
28
|
Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23063346. [PMID: 35328769 PMCID: PMC8954705 DOI: 10.3390/ijms23063346] [Citation(s) in RCA: 257] [Impact Index Per Article: 128.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with these processes, results in cardiovascular complications. This review focuses on the different stages of atherosclerosis development, ranging from endothelial dysfunction to plaque rupture. In addition, the post-transcriptional regulation and modulation of atheroma plaque by microRNAs and lncRNAs, the role of microbiota, and the importance of sex as a crucial risk factor in atherosclerosis are covered here in order to provide a global view of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
| | - Unai Galicia-García
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | | | - Iraide Alloza
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Koen Vandenbroeck
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Bizkaia, Spain
| | - Asier Benito-Vicente
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| | - César Martín
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| |
Collapse
|
29
|
Maheshwari U, Huang SF, Sridhar S, Keller A. The Interplay Between Brain Vascular Calcification and Microglia. Front Aging Neurosci 2022; 14:848495. [PMID: 35309892 PMCID: PMC8924545 DOI: 10.3389/fnagi.2022.848495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/11/2022] [Indexed: 12/17/2022] Open
Abstract
Vascular calcifications are characterized by the ectopic deposition of calcium and phosphate in the vascular lumen or wall. They are a common finding in computed tomography scans or during autopsy and are often directly related to a pathological condition. While the pathogenesis and functional consequences of vascular calcifications have been intensively studied in some peripheral organs, vascular calcification, and its pathogenesis in the central nervous system is poorly characterized and understood. Here, we review the occurrence of vessel calcifications in the brain in the context of aging and various brain diseases. We discuss the pathomechanism of brain vascular calcification in primary familial brain calcification as an example of brain vessel calcification. A particular focus is the response of microglia to the vessel calcification in the brain and their role in the clearance of calcifications.
Collapse
Affiliation(s)
- Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
| | - Sucheta Sridhar
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zurich, Switzerland
- *Correspondence: Annika Keller,
| |
Collapse
|
30
|
Gnemmi V, Li Q, Ma Q, De Chiara L, Carangelo G, Li C, Molina-Van den Bosch M, Romagnani P, Anders HJ, Steiger S. Asymptomatic Hyperuricemia Promotes Recovery from Ischemic Organ Injury by Modulating the Phenotype of Macrophages. Cells 2022; 11:cells11040626. [PMID: 35203277 PMCID: PMC8869798 DOI: 10.3390/cells11040626] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Acute organ injury, such as acute kidney injury (AKI) and disease (AKD), are major causes of morbidity and mortality worldwide. Hyperuricemia (HU) is common in patients with impaired kidney function but the impact of asymptomatic HU on the different phases of AKI/AKD is incompletely understood. We hypothesized that asymptomatic HU would attenuate AKD because soluble, in contrast to crystalline, uric acid (sUA) can attenuate sterile inflammation. In vitro, 10 mg/dL sUA decreased reactive oxygen species and interleukin-6 production in macrophages, while enhancing fatty acid oxidation as compared with a physiological concentration of 5 mg/dL sUA or medium. In transgenic mice, asymptomatic HU of 7–10 mg/dL did not affect post-ischemic AKI/AKD but accelerated the recovery of kidney excretory function on day 14. Improved functional outcome was associated with better tubular integrity, less peritubular inflammation, and interstitial fibrosis. Mechanistic studies suggested that HU shifted macrophage polarization towards an anti-inflammatory M2-like phenotype characterized by expression of anti-oxidative and metabolic genes as compared with post-ischemic AKI-chronic kidney disease transition in mice without HU. Our data imply that asymptomatic HU acts as anti-oxidant on macrophages and tubular epithelial cells, which endorses the recovery of kidney function and structure upon AKI.
Collapse
Affiliation(s)
- Viviane Gnemmi
- Service d’Anatomie Pathologique, Centre de Biologie Pathologique, CHU Lille, 59037 Lille, France;
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Qiubo Li
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Letizia De Chiara
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (L.D.C.); (G.C.); (P.R.)
| | - Giulia Carangelo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (L.D.C.); (G.C.); (P.R.)
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Mireia Molina-Van den Bosch
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Paola Romagnani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (L.D.C.); (G.C.); (P.R.)
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (Q.L.); (Q.M.); (C.L.); (M.M.-V.d.B.); (H.-J.A.)
- Correspondence:
| |
Collapse
|
31
|
Elmonem MA, Veys KRP, Prencipe G. Nephropathic Cystinosis: Pathogenic Roles of Inflammation and Potential for New Therapies. Cells 2022; 11:cells11020190. [PMID: 35053306 PMCID: PMC8773784 DOI: 10.3390/cells11020190] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 01/18/2023] Open
Abstract
The activation of several inflammatory pathways has recently been documented in patients and different cellular and animal models of nephropathic cystinosis. Upregulated inflammatory signals interact with many pathogenic aspects of the disease, such as enhanced oxidative stress, abnormal autophagy, inflammatory cell recruitment, enhanced cell death, and tissue fibrosis. Cysteamine, the only approved specific therapy for cystinosis, ameliorates many but not all pathogenic aspects of the disease. In the current review, we summarize the inflammatory mechanisms involved in cystinosis and their potential impact on the disease pathogenesis and progression. We further elaborate on the crosstalk between inflammation, autophagy, and apoptosis, and discuss the potential of experimental drugs for suppressing the inflammatory signals in cystinosis.
Collapse
Affiliation(s)
- Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 11628, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
- Correspondence:
| | - Koenraad R. P. Veys
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Department of Pediatrics, AZ Delta Campus, 8820 Torhout, Belgium
| | - Giusi Prencipe
- Laboratory of Immuno-Rheumatology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| |
Collapse
|
32
|
Favor OK, Pestka JJ, Bates MA, Lee KSS. Centrality of Myeloid-Lineage Phagocytes in Particle-Triggered Inflammation and Autoimmunity. FRONTIERS IN TOXICOLOGY 2021; 3:777768. [PMID: 35295146 PMCID: PMC8915915 DOI: 10.3389/ftox.2021.777768] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Exposure to exogenous particles found as airborne contaminants or endogenous particles that form by crystallization of certain nutrients can activate inflammatory pathways and potentially accelerate autoimmunity onset and progression in genetically predisposed individuals. The first line of innate immunological defense against particles are myeloid-lineage phagocytes, namely macrophages and neutrophils, which recognize/internalize the particles, release inflammatory mediators, undergo programmed/unprogrammed death, and recruit/activate other leukocytes to clear the particles and resolve inflammation. However, immunogenic cell death and release of damage-associated molecules, collectively referred to as "danger signals," coupled with failure to efficiently clear dead/dying cells, can elicit unresolved inflammation, accumulation of self-antigens, and adaptive leukocyte recruitment/activation. Collectively, these events can promote loss of immunological self-tolerance and onset/progression of autoimmunity. This review discusses critical molecular mechanisms by which exogenous particles (i.e., silica, asbestos, carbon nanotubes, titanium dioxide, aluminum-containing salts) and endogenous particles (i.e., monosodium urate, cholesterol crystals, calcium-containing salts) may promote unresolved inflammation and autoimmunity by inducing toxic responses in myeloid-lineage phagocytes with emphases on inflammasome activation and necrotic and programmed cell death pathways. A prototypical example is occupational exposure to respirable crystalline silica, which is etiologically linked to systemic lupus erythematosus (SLE) and other human autoimmune diseases. Importantly, airway instillation of SLE-prone mice with crystalline silica elicits severe pulmonary pathology involving accumulation of particle-laden alveolar macrophages, dying and dead cells, nuclear and cytoplasmic debris, and neutrophilic inflammation that drive cytokine, chemokine, and interferon-regulated gene expression. Silica-induced immunogenic cell death and danger signal release triggers accumulation of T and B cells, along with IgG-secreting plasma cells, indicative of ectopic lymphoid tissue neogenesis, and broad-spectrum autoantibody production in the lung. These events drive early autoimmunity onset and accelerate end-stage autoimmune glomerulonephritis. Intriguingly, dietary supplementation with ω-3 fatty acids have been demonstrated to be an intervention against silica-triggered murine autoimmunity. Taken together, further insight into how particles drive immunogenic cell death and danger signaling in myeloid-lineage phagocytes and how these responses are influenced by the genome will be essential for identification of novel interventions for preventing and treating inflammatory and autoimmune diseases associated with these agents.
Collapse
Affiliation(s)
- Olivia K. Favor
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Melissa A. Bates
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
33
|
Baumer Y, McCurdy SG, Boisvert WA. Formation and Cellular Impact of Cholesterol Crystals in Health and Disease. Adv Biol (Weinh) 2021; 5:e2100638. [PMID: 34590446 PMCID: PMC11055929 DOI: 10.1002/adbi.202100638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/20/2021] [Indexed: 11/10/2022]
Abstract
Cholesterol crystals (CCs) were first discovered in atherosclerotic plaque tissue in the early 1900 and have since been observed and implicated in many diseases and conditions, including myocardial infarction, abdominal aortic aneurism, kidney disease, ocular diseases, and even central nervous system anomalies. Despite the widespread involvement of CCs in many pathologies, the mechanisms involved in their formation and their role in various diseases are still not fully understood. Current knowledge concerning the formation of CCs, as well as the molecular pathways activated upon cellular exposure to CCs, will be explored in this review. As CC formation is tightly associated with lipid metabolism, the role of cellular lipid homeostasis in the formation of CCs is highlighted, including the role of lysosomes. In addition, cellular pathways and processes known to be affected by CCs are described. In particular, CC-induced activation of the inflammasome and production of reactive oxygen species, along with the role of CCs in complement-mediated inflammation is discussed. Moreover, the clinical manifestation of embolized CCs is described with a focus on renal and skin diseases associated with CC embolism. Lastly, potential therapeutic measures that target either the formation of CCs or their impact on different cell types and tissues are highlighted.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung, and Blood Institute, Building 10, 10 Center Drive, Bethesda, MD 20814, USA
| | - Sara G. McCurdy
- Dept. of Medicine, University of California San Diego, 9500 Gilman Street, La Jolla, CA 92093, USA
| | - William A. Boisvert
- Center for Cardiovascular Research, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, USA
| |
Collapse
|
34
|
Gelardi M, Netti GS, Giancaspro R, Spadaccino F, Pennella A, Fiore V, La Gatta E, Grilli GM, Cassano M, Ranieri E. Chronic rhinosinusitis with nasal polyposis (CRSwNP): the correlation between expression of Galectin-10 and Clinical-Cytological Grading (CCG). Am J Rhinol Allergy 2021; 36:229-237. [PMID: 34647485 DOI: 10.1177/19458924211049867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is typically characterized by Type 2 inflammation. Several biomarkers of eosinophilic inflammation, including Galectin-10, also known as Charcot-Leyden crystal protein (CLCP), have been identified to establish eosinophilic infiltration of polyps, a reliable predictor of recurrence.Objective: We aimed to evaluate the Galectin-10 expression in nasal polyps of patients with CRSwNP and to assess the correlation of Charcot-Leyden crystals expression to the severity of CRSwNP according to Clinical-Cytological Grading (CCG). METHODS A double-label immunofluorescence was performed to evaluate the expression of Gal-10, CD15, Tryptase, and CD63 and their eventual co-localization on histological samples of 18 patients with CRSwNP. Double-positive Gal-10+CD15+ and Galectin-10+Tryptase+ inflammatory cells were counted by confocal microscopy. RESULTS Galectin-10 was detectable in all examined tissues from CRSwNP patients, and its expression increased as low, medium and high CCG tissues were examined, respectively. Galectin-10 was extensively present in inflammatory cells, while limited Galectin-10 deposits were detected around mucosal epithelial cells. CONCLUSION We showed the strong correlation between CCG and Galectin-10 expression, mainly colocalized with infiltrating eosinophils and mast-cells, in patients affected by CRSwNP.
Collapse
Affiliation(s)
- Matteo Gelardi
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Giuseppe Stefano Netti
- Unit of Clinical Pathology and Center for Molecular Medicine, 507873Department of Medical and Surgical Sciences, 18972University of Foggia, Foggia, Italy
| | - Rossana Giancaspro
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Federica Spadaccino
- Unit of Clinical Pathology and Center for Molecular Medicine, 507873Department of Medical and Surgical Sciences, 18972University of Foggia, Foggia, Italy
| | - Antonio Pennella
- Unit of Pathology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Valeria Fiore
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Emanuel La Gatta
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Gianluigi Mariano Grilli
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Michele Cassano
- Unit of Otolaryngology, Department of Clinical and Experimental Medicine, 18972University of Foggia, Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology and Center for Molecular Medicine, 507873Department of Medical and Surgical Sciences, 18972University of Foggia, Foggia, Italy
| |
Collapse
|
35
|
Kuroda A. Recent progress and perspectives on the mechanisms underlying Asbestos toxicity. Genes Environ 2021; 43:46. [PMID: 34641979 PMCID: PMC8507173 DOI: 10.1186/s41021-021-00215-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/13/2021] [Indexed: 01/10/2023] Open
Abstract
Most cases of mesothelioma are known to result from exposure to asbestos fibers in the environment or occupational ambient air. The following questions regarding asbestos toxicity remain partially unanswered: (i) why asbestos entering the alveoli during respiration exerts toxicity in the pleura; and (ii) how asbestos causes mesothelioma, even though human mesothelial cells are easily killed upon exposure to asbestos. As for the latter question, it is now thought that the frustrated phagocytosis of asbestos fibers by macrophages prolongs inflammatory responses and gives rise to a “mutagenic microenvironment” around mesothelial cells, resulting in their malignant transformation. Based on epidemiological and genetic studies, a carcinogenic model has been proposed in which BRCA1-associated protein 1 mutations are able to suppress cell death in mesothelial cells and increase genomic instability in the mutagenic microenvironment. This leads to additional mutations, such as CDKN2A [p16], NF2, TP53, LATS2, and SETD2, which are associated with mesothelioma carcinogenesis. Regarding the former question, the receptors involved in the intracellular uptake of asbestos and the mechanism of transfer of inhaled asbestos from the alveoli to the pleura are yet to be elucidated. Further studies using live-cell imaging techniques will be critical to fully understanding the mechanisms underlying asbestos toxicity.
Collapse
Affiliation(s)
- Akio Kuroda
- Unit of Biotechnology, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530, Japan.
| |
Collapse
|
36
|
Bouabdallaoui N, Tardif JC. Colchicine in the Management of Acute and Chronic Coronary Artery Disease. Curr Cardiol Rep 2021; 23:120. [PMID: 34269908 DOI: 10.1007/s11886-021-01560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Inflammation is involved in the initiation, progression, and destabilization of atherosclerosis. Anti-inflammatory strategies aimed at reducing residual cardiovascular (CV) risk have gained increasing interest in addition to the traditional management of risk factors. Colchicine is a potent anti-inflammatory therapy that affects the inflammasome and other targets. We will herein review the most recent evidence regarding the usefulness of colchicine in patients with coronary artery disease (CAD). RECENT FINDINGS Colchicine has recently been repurposed from its traditional use to a number of CV indications. The landmark COLCOT and LoDoCo2 trials have demonstrated that long-term use of colchicine was associated with a reduced rate of CV events in both acute and chronic presentations of CAD, with an overall good safety profile. Colchicine is emerging as a valuable, safe, and cost-effective therapy in addition to standard of care for the prevention of atherothrombotic events in CAD.
Collapse
Affiliation(s)
- Nadia Bouabdallaoui
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec, H1T1C8, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec, H1T1C8, Canada.
| |
Collapse
|
37
|
Galozzi P, Bindoli S, Luisetto R, Sfriso P, Ramonda R, Scanu A, Oliviero F. Regulation of crystal induced inflammation: current understandings and clinical implications. Expert Rev Clin Immunol 2021; 17:773-787. [PMID: 34053376 DOI: 10.1080/1744666x.2021.1937129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Accumulation of abnormal crystals in the body, derived from endogenous or exogenous materials can drive a wide spectrum of inflammatory disease states. It is well established that intra-articular deposition of monosodium urate (MSU) and calcium pyrophoshate (CPP) crystals contributes to joint destruction through pro-inflammatory processes.Areas covered: This review will focus on current understanding and recent novelty about the mechanisms and the clinical implications of the inflammation induced by MSU and CPP crystals.Expert opinion: Advances in molecular biology reveal that at the base of the inflammatory cascade, stimulated by MSU or CPP crystals, there are many complex cellular mechanisms mainly involving the NLRP3 inflammasome, the hallmark of autoinflammatory syndromes. The extensive studies carried out through in vitro and in vivo models along with a better clinical definition of the disease has led to an optimized use of existing drugs and the introduction of novel therapeutic strategies. In particular, the identification of IL-1 as the most important target in gout and pseudogout has made it possible to expand the pharmacological indications of anti-IL-1 biological drugs, opening new therapeutic perspectives for patients.
Collapse
Affiliation(s)
- Paola Galozzi
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Sara Bindoli
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padova, Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Anna Scanu
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| |
Collapse
|
38
|
Matassa R, Cattaruzza MS, Sandorfi F, Battaglione E, Relucenti M, Familiari G. Direct imaging evidences of metal inorganic contaminants traced into cigarettes. JOURNAL OF HAZARDOUS MATERIALS 2021; 411:125092. [PMID: 33858086 DOI: 10.1016/j.jhazmat.2021.125092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/20/2020] [Accepted: 01/06/2021] [Indexed: 05/14/2023]
Abstract
Today, environmental health research on toxicological adverse effects of metal-inorganic materials diffused by cigarettes represents a new challenge for assessing new health risks directly related to the critical chemical-size features of the particles. Therefore, morpho-chemical analyses of hazardous particles become critical in response to the distinctive assumptions about the origin, evolution, and coexisting phases. Here, we report a detailed investigation through direct microscopy imaging of metal-inorganic contaminants for one traditional and two heat-not-burn commercial cigarettes of three different brands. Chemical-size studies revealed the critical presence of heavy metal-inorganic nanostructured microparticles on both paper and filter components of the cigarette, before and after smoking. The direct experimental imaging evidenced on how hazardous particles evolved in mass-size forming coexisting multi-phases of large agglomerate because of the persistence and accumulative effect of the heating puffing. The estimated porosity of the unsuitable engineered filters validated the allowed migration of micrometric pollutants independently from their intrinsic size-shape property. Furthermore, the inappropriate design of the filters made it an adverse sponge reservoir capable of collecting all possible hazardous chemical agents potentially toxic. These substantial results strongly support experimentally the tremendous effect of the smoke capable of transporting and manipulating a high amount of elusive particles, as a particles heat carrier.
Collapse
Affiliation(s)
- Roberto Matassa
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy.
| | - Maria Sofia Cattaruzza
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Filippo Sandorfi
- Department of Hygiene and Public Health, Alma Mater Studiorum University of Bologna, via San Giacomo 12, 40126 Bologna, Italy
| | - Ezio Battaglione
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy
| | - Giuseppe Familiari
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy
| |
Collapse
|
39
|
Flores E, Sánchez-Sánchez M, Gutierrez C, Estébanez B, Millán P, Gutierrez C, Gonzalez A, Garcia de Lorenzo A. High Dose Ascorbic Acid During Acute Resuscitation in Critically Burn Patients. J Burn Care Res 2021; 43:149-155. [PMID: 34142146 DOI: 10.1093/jbcr/irab088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Ascorbic acid (AA) is a potent oxygen-free radical scavenger. We hypothesized that treating severe burn patients with high doses of AA (HDAA) can reduce fluid resuscitation requirements and prevent organ dysfunction. We performed a unicentric, retrospective case-control study of 75 burn patients: 25 patients admitted from 2018 to 2019 with more than 30% Total Surface Body Surface Area (TSBA) burned who received HDAA (66 mg/kg/h as soon as possible after admission until 36 h after injury), and 50 patients admitted from 2014 to 2017 with similar Abbreviated Burn Severity Index (ABSI)/Baux scores who were treated with the same protocol but did not receive HDAA. During the first 24 hours of burn resuscitation the HDAA group required less fluids than the control group (3.06 ± 0.87 ml/kg/%TBSA vs 4.32 ± 1.51 P < .05), but the overall reduction of fluid requirements during the first 72 hours was not significant. There were no significant differences in Sequential Organ Failure Assessment (SOFA), other hemodynamic parameters, complications, or mortality. We also did not find an increase acute kidney injury in patients who received HDAA even though the mean urine oxalate/creatinine ratio was 0.61 (0.02-0.96). We conclude that in severe burn patients treated with a restrictive fluid therapy protocol, administration of HDAA can decrease only the initial fluid requirements but not total fluid intakes. We did not find differences in severity score after resuscitation or in mortality. Nor did we find an increase in renal failure in patients administered with HDAA.
Collapse
Affiliation(s)
- Eva Flores
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Manuel Sánchez-Sánchez
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Claudio Gutierrez
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Belen Estébanez
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Pablo Millán
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Carola Gutierrez
- Department of Intensive Care Medicine, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | - Alvaro Gonzalez
- Department of Plastic and Reconstructive Surgery, University Hospital La Paz-Carlos III/IdiPAZ, Madrid, Spain
| | | |
Collapse
|
40
|
Stallings TL, Riefkohl Lisci A, McCray NL, Weiner DE, Kaufman JS, Aschengrau A, Ma Y, LaValley MP, Ramírez-Rubio O, Jose Amador J, López-Pilarte D, Laws RL, Winter M, McSorley VE, Brooks DR, Applebaum KM. Dysuria, heat stress, and muscle injury among Nicaraguan sugarcane workers at risk for Mesoamerican nephropathy. Scand J Work Environ Health 2021; 47:377-386. [PMID: 34003295 PMCID: PMC8259701 DOI: 10.5271/sjweh.3963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Objectives: Nicaraguan sugarcane workers, particularly cane cutters, have an elevated prevalence of chronic kidney disease of unknown origin, also referred to as Mesoamerican nephropathy (MeN). The pathogenesis of MeN may include recurrent heat stress, crystalluria, and muscle injury with subsequent kidney injury. Yet, studies examining the frequency of such events in long-term, longitudinal studies are limited. Methods: Using employment and medical data for male workers at a Nicaraguan sugarcane company, we classified months of active work as either work as a cane cutter or other sugarcane job and determined occurrence of dysuria, heat events and muscle events. Work months and events occurred January 1997 to June 2010. Associations between cane cutting and each outcome were analyzed using logistic regression based on generalized estimating equations for repeated events, controlling for age. Results: Among 242 workers with 7257 active work months, 19.5% of person-months were as a cane cutter. There were 160, 21, and 16 episodes of dysuria, heat events, and muscle events, respectively. Compared with work months in other jobs, cane cutting was associated with an elevated odds of dysuria [odds ratio 2.40 (95% confidence interval 1.56–3.68)]. The number of heat and muscle events by cane cutter and other job were limited. Conclusions: Working as a cane cutter compared with other jobs in the sugarcane industry was associated with increased dysuria, supporting the hypothesis that cane cutters are at increased risk of events suspected of inducing or presaging clinically evident kidney injury.
Collapse
Affiliation(s)
- Tiffany L Stallings
- The George Washington University, Milken Institute School of Public Health, Department of Environmental and Occupational Health, 950 New Hampshire Ave., NW, Suite 400, Washington, DC 20052, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tantranont N, Luque Y, Hsiao M, Haute C, Gaber L, Barrios R, Adrogue HE, Niasse A, Truong LD. Vancomycin-Associated Tubular Casts and Vancomycin Nephrotoxicity. Kidney Int Rep 2021; 6:1912-1922. [PMID: 34307986 PMCID: PMC8258501 DOI: 10.1016/j.ekir.2021.04.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Introduction Vancomycin nephrotoxicity is frequent and may be due to drug-induced acute tubular necrosis (ATN) or tubulointerstitial nephritis (TIN). Vancomycin-associated tubular cast (VTC) was recently described and may represent a novel cause of vancomycin nephrotoxicity. However, much is still unknown about VTC. Materials and Methods Thirty-seven kidney biopsy specimens from patients who were treated with vancomycin and developed acute kidney injury (AKI) were found among a total of 4673 biopsy samples between 2010 and 2019. These biopsy specimens were subjected to light microscopy, immunofluorescence, electron microscopy, and immunolocalization for vancomycin, uromodulin, myoglobin, tubular segment–specific markers, and examined for VTCs. The findings were correlated with the clinical course. Results VTCs displayed precipitated vancomycin casts in a background of uromodulin; the casts were limited to the distal tubules, and always associated with a background of more diffuse renal injury (ATN or TIN). The diagnosis of vancomycin nephrotoxicity was made in in 28 of 37 patients. VTC was noted in 25 of 28 biopsy samples from patients diagnosed with vancomycin nephrotoxicity and in one of nine biopsy samples from patients without this diagnosis. Vancomycin nephrotoxicity was diagnosed in 25 of 26 patients whose biopsy specimens showed VTC, but in only 3 of 11 patients without VTC in the biopsy samples. Conclusions VTC displays a characteristic morphologic profile amenable to ready recognition in biopsy specimens. It results from coprecipitation of vancomycin and uromodulin. It facilitates the biopsy diagnosis of vancomycin nephrotoxicity. It may have a nephrotoxic effect superimposing on and independent from the ATN or interstitial nephritis in the pathogenesis of vancomycin nephrotoxicity.
Collapse
Affiliation(s)
- Ngoentra Tantranont
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA.,Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yosu Luque
- Soins Intensifs Néphrologiques et Rein Aigu, Tenon Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Inserm UMR_S1155, Sorbonne Université, Paris, France
| | - Mary Hsiao
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Claire Haute
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Lillian Gaber
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Roberto Barrios
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Horacio E Adrogue
- Department of Medicine, Renal Section, Houston Methodist Hospital, Houston, Texas, USA
| | | | - Luan D Truong
- Department of Pathology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| |
Collapse
|
42
|
Abstract
In the last decade, the role of apoptosis in the pathophysiology of acute kidney injury (AKI) and AKI to chronic kidney disease (CKD) progression has been revisited as our understanding of ferroptosis and necroptosis has emerged. A growing body of evidence, reviewed here, ascribes a central pathophysiological role for ferroptosis and necroptosis to AKI, nephron loss, and acute tubular necrosis. We will introduce concepts to the non-cell-autonomous manner of kidney tubular injury during ferroptosis, a phenomenon that we refer to as a "wave of death." We hypothesize that necroptosis might initiate cell death propagation through ferroptosis. The remaining necrotic debris requires effective removal processes to prevent a secondary inflammatory response, referred to as necroinflammation. Open questions include the differences in the immunogenicity of ferroptosis and necroptosis, and the specificity of necrostatins and ferrostatins to therapeutically target these processes to prevent AKI-to-CKD progression and end-stage renal disease.
Collapse
|
43
|
Hannan M, Ansari S, Meza N, Anderson AH, Srivastava A, Waikar S, Charleston J, Weir MR, Taliercio J, Horwitz E, Saunders MR, Wolfrum K, Feldman HI, Lash JP, Ricardo AC. Risk Factors for CKD Progression: Overview of Findings from the CRIC Study. Clin J Am Soc Nephrol 2021; 16:648-659. [PMID: 33177074 PMCID: PMC8092061 DOI: 10.2215/cjn.07830520] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Chronic Renal Insufficiency Cohort (CRIC) Study is an ongoing, multicenter, longitudinal study of nearly 5500 adults with CKD in the United States. Over the past 10 years, the CRIC Study has made significant contributions to the understanding of factors associated with CKD progression. This review summarizes findings from longitudinal studies evaluating risk factors associated with CKD progression in the CRIC Study, grouped into the following six thematic categories: (1) sociodemographic and economic (sex, race/ethnicity, and nephrology care); (2) behavioral (healthy lifestyle, diet, and sleep); (3) genetic (apoL1, genome-wide association study, and renin-angiotensin-aldosterone system pathway genes); (4) cardiovascular (atrial fibrillation, hypertension, and vascular stiffness); (5) metabolic (fibroblast growth factor 23 and urinary oxalate); and (6) novel factors (AKI and biomarkers of kidney injury). Additionally, we highlight areas where future research is needed, and opportunities for interdisciplinary collaboration.
Collapse
Affiliation(s)
- Mary Hannan
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Sajid Ansari
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Natalie Meza
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Amanda H. Anderson
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Anand Srivastava
- Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Sushrut Waikar
- Nephrology Section, Boston University Medical Center, Boston, Massachusetts
| | - Jeanne Charleston
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Matthew R. Weir
- Division of Nephrology, Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Jonathan Taliercio
- Department of Nephrology and Hypertension, Cleveland Clinic, Cleveland, Ohio
| | | | - Milda R. Saunders
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Katherine Wolfrum
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Harold I. Feldman
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James P. Lash
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Ana C. Ricardo
- Department of Medicine, University of Illinois, Chicago, Illinois
| | | |
Collapse
|
44
|
Feurle GE, Moos V, Stroux A, Gehrmann-Sommer N, Poddubnyy D, Fiehn C, Schneider T. Differential diagnostic value of rheumatic symptoms in patients with Whipple's disease. Sci Rep 2021; 11:5980. [PMID: 33727566 PMCID: PMC7966399 DOI: 10.1038/s41598-021-85217-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Most patients with Whipple’s disease have rheumatic symptoms. The aim of our prospective, questionnaire-based, non-interventional clinical study was to assess whether these symptoms are useful in guiding the differential diagnosis to the rheumatic disorders. Forty patients with Whipple’s disease, followed by 20 patients for validation and 30 patients with rheumatoid-, 21 with psoriatic-, 15 with palindromic- and 25 with axial spondyloarthritis were recruited for the present investigation. Patients with Whipple’s disease and patients with rheumatic disorders were asked to record rheumatic symptoms on pseudonymized questionnaires. The data obtained were subjected to multiple logistic regression analysis. Episodic pain with rapid onset, springing from joint to joint was most common in patients with palindromic arthritis and second most common and somewhat less conspicuous in Whipple’s disease. Continuous pain in the same joints predominated in patients with rheumatoid-, psoriatic-, and axial spondyloarthritis. Multiple logistic equations resulted in a predicted probability for the diagnosis of Whipple’s disease of 43.4 ± 0.19% (M ± SD) versus a significantly lower probability of 23.8 ± 0.19% (M ± SD) in the aggregate of patients with rheumatic disorders. Mean area under the curve (AUC) ± SD was 0.781 ± 0.044, 95% CI 0.695–0.867, asymptotic significance p < 0.001. The logistic equations predicted probability for the diagnosis of Whipple’s disease in the initial series of 40 patients of 43.4 ± 0.19% was not significantly different in the subsequent 20 patients of 38.2 ± 0.28% (M ± SD) (p = 0.376). The data may be useful in a predictive algorithm for diagnosing Whipple’s disease. The project is registered as clinical study DRK S0001566.
Collapse
Affiliation(s)
- Gerhard E Feurle
- DRK Krankenhaus Neuwied, Eduard Moerikestrasse 12, 56567, Neuwied, Germany.
| | - Verena Moos
- Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Stroux
- Institut für Biometrie und Klinische Epidemiologie, Charité-Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Nadine Gehrmann-Sommer
- Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Fiehn
- Praxis für Rheumatologie, Klinische Immunologie, Medical Center, Baden-Baden, Germany
| | - Thomas Schneider
- Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
45
|
Prajapati S, Tomar B, Srivastava A, Narkhede YB, Gaikwad AN, Lahiri A, Mulay SR. 6,7-Dihydroxycoumarin ameliorates crystal-induced necroptosis during crystal nephropathies by inhibiting MLKL phosphorylation. Life Sci 2021; 271:119193. [PMID: 33577856 DOI: 10.1016/j.lfs.2021.119193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 01/09/2023]
Abstract
AIMS Mineralization of crystalline particles and the formation of renal calculi contribute to the pathogenesis of crystal nephropathies. Several recent studies on the biology of crystal handling implicated intrarenal crystal deposition-induced necroinflammation in their pathogenesis. We hypothesized that 6,7-dihydroxycoumarin (DHC) inhibit intrarenal crystal cytotoxicity and necroinflammation, and ameliorate crystal-induced chronic kidney disease (CKD). MAIN METHODS An unbiased high content screening coupled with fluorescence microscopy was used to identify compounds that inhibit CaOx crystal cytotoxicity. The ligand-protein interactions were identified using computational models e.g. molecular docking and molecular dynamics simulations. Furthermore, mice and rat models of oxalate-induced CKD were used for in-vivo studies. Renal injury, crystal deposition, and fibrosis were assessed by histology analysis. Western blots were used to quantify the protein expression. Data were expressed as boxplots and analyzed using one way ANOVA. KEY FINDINGS An unbiased high-content screening in-vitro identified 6,7-DHC as a promising candidate. Further, 6,7-DHC protected human and mouse cells from calcium oxalate (CaOx) crystal-induced necroptosis in-vitro as well as mice and rats from oxalate-induced CKD in either preventive or therapeutic manner. Computational modeling demonstrated that 6,7-DHC interact with MLKL, the key protein in the necroptosis machinery, and inhibit its phosphorylation by ATP, which was evident in both in-vitro and in-vivo analyses. SIGNIFICANCE Together, our results indicate that 6,7-DHC possesses a novel pharmacological property as a MLKL inhibitor and could serve as a lead molecule for further development of coumarin-based novel MLKL inhibitors. Furthermore, our data identify 6,7-DHC as a novel therapeutic strategy to combat crystal nephropathies.
Collapse
Affiliation(s)
- Smita Prajapati
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Bhawna Tomar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anjali Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Yogesh B Narkhede
- Department of Bioengineering, Bourn's College of Engineering, University of California, CA 92521, USA
| | - Anil N Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit Lahiri
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shrikant R Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
46
|
Omori S, Tsugita M, Hoshikawa Y, Morita M, Ito F, Yamaguchi SI, Xie Q, Noyori O, Yamaguchi T, Takada A, Saitoh T, Toyokuni S, Akiba H, Nagata S, Kinoshita K, Nakayama M. Tim4 recognizes carbon nanotubes and mediates phagocytosis leading to granuloma formation. Cell Rep 2021; 34:108734. [PMID: 33567275 DOI: 10.1016/j.celrep.2021.108734] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/02/2020] [Accepted: 01/19/2021] [Indexed: 12/22/2022] Open
Abstract
Macrophage recognition and phagocytosis of crystals is critical for the associated fibrosis and cancer. Of note, multi-walled carbon nanotubes (MWCNTs), the highly representative products of nanotechnology, induce macrophage NLRP3 inflammasome activation and cause asbestosis-like pathogenesis. However, it remains largely unknown how macrophages efficiently recognize MWCNTs on their cell surfaces. Here, we identify by a targeted screening of phagocyte receptors the phosphatidylserine receptors T cell immunoglobulin mucin 4 (Tim4) and Tim1 as the pattern-recognition receptors for carbon crystals. Docking simulation studies reveal spatiotemporally stable interfaces between aromatic residues in the extracellular IgV domain of Tim4 and one-dimensional carbon crystals. Further, CRISPR-Cas9-mediated deletion of Tim4 and Tim1 reveals that Tim4, but not Tim1, critically contributes to the recognition of MWCNTs by peritoneal macrophages and to granuloma development in a mouse model of direct mesothelium exposure to MWCNTs. These results suggest that Tim4 recognizes MWCNTs through aromatic interactions and mediates phagocytosis leading to granulomas.
Collapse
Affiliation(s)
- Satoshi Omori
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Misato Tsugita
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Yasuto Hoshikawa
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumiya Ito
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan
| | - Shin-Ichiro Yamaguchi
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Qilin Xie
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Osamu Noyori
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Tomoya Yamaguchi
- Department of Cancer Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; PRESTO, JST, Kawaguchi, Japan
| | - Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Hisaya Akiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kengo Kinoshita
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.
| | - Masafumi Nakayama
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan; Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan; PRESTO, JST, Kawaguchi, Japan.
| |
Collapse
|
47
|
Alberts A, Klingberg A, Hoffmeister L, Wessig AK, Brand K, Pich A, Neumann K. Binding of Macrophage Receptor MARCO, LDL, and LDLR to Disease-Associated Crystalline Structures. Front Immunol 2020; 11:596103. [PMID: 33363539 PMCID: PMC7753766 DOI: 10.3389/fimmu.2020.596103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Endogenous and exogenous crystalline structures are involved in various pathologies and diseases in humans by inducing sterile inflammation, mechanical stress, or obstruction of excretory organs. The best studied of these diseases is gout, in which crystallization of uric acid in the form of monosodium urate (MSU) mainly in synovial fluid of the joints leads to sterile inflammation. Though some of these diseases have been described for centuries, little is known about if and how the immune system recognizes the associated crystals. Thus, in this study we aimed at identifying possible recognition molecules of MSU using liquid chromatography-mass spectrometry (LC-MS) analysis of MSU-binding serum proteins. Among the strongest binding proteins, we unexpectedly found two transmembrane receptors, namely macrophage receptor with collagenous structure (MARCO) and low-density lipoprotein (LDL) receptor (LDLR). We show that recombinant versions of both human and mouse MARCO directly bind to unopsonized MSU and several other disease-associated crystals. Recombinant LDLR binds many types of crystals mainly when opsonized with serum proteins. We show that this interaction is predominantly mediated by LDL, which we found to bind to all crystalline structures tested except for cholesterol crystals. However, murine macrophages lacking LDLR expression do neither show altered phagocytosis nor interleukin-1β (IL-1β) production in response to opsonized crystals. Binding of LDL to MSU has previously been shown to inhibit the production of reactive oxygen species (ROS) by human neutrophils. We extend these findings and show that LDL inhibits neutrophil ROS production in response to most crystals tested, even cholesterol crystals. The inhibition of neutrophil ROS production only partly correlated with the inhibition of IL-1β production by peripheral blood mononuclear cells (PBMCs): LDL inhibited IL-1β production in response to large MSU crystals, but not small MSU or silica crystals. This may suggest distinct upstream signals for IL-1β production depending on the size or the shape of the crystals. Together, we identify MARCO and LDLR as potential crystal recognition receptors, and show that LDL binding to diverse disease-associated crystalline structures has variable effects on crystal-induced innate immune cell activation.
Collapse
Affiliation(s)
- Anika Alberts
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Annika Klingberg
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Leonie Hoffmeister
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Korbinian Brand
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Andreas Pich
- Research Core Unit Proteomics & Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Konstantin Neumann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
48
|
Kim T, de Oliveira Silva Lautenschlager S, Ma Q, Eller K, Pollheimer MJ, Lazarin-Bidóia D, Nakamura CV, Anders HJ, Steiger S. Drug Crystal-Related Gastrointestinal Complications Involve Crystal-Induced Release of Neutrophil and Monocyte Extracellular Traps. Cells 2020; 9:cells9112481. [PMID: 33203124 PMCID: PMC7697008 DOI: 10.3390/cells9112481] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022] Open
Abstract
Ion-exchange resins are commonly used to manage complications of chronic kidney disease, such as hyperphosphatemia, hyperkalemia, and hypercholesterolemia. Occasionally, these drugs can irritate the gastrointestinal lining and cause life-threatening intestinal necrosis. Currently, the pathophysiology of drug crystal-induced intestinal necrosis is not well understood. We hypothesized that crystals of ion-exchange resins like sevelamer, polystyrene sulfonate, and cholestyramine can trigger the formation of neutrophil and monocyte extracellular traps by contributing to intestinal barrier dysfunction. Light and fluorescence microscopy of the colonic resection specimen from a patient with chronic kidney disease revealed severe intestinal necrosis, ulceration, sevelamer crystals, and inflammation upon oral intake of sevelamer, as well as the formation of neutrophil extracellular traps in proximity to small sevelamer crystals. Indeed, drug crystals reduced metabolic activity and induced barrier dysfunction and cell death in human intestinal epithelial cells in vitro. In addition, drug crystals triggered the release of neutrophil and monocyte extracellular traps. Taken together, these data raise the possibility that besides other factors including chronic kidney disease, diabetes mellitus, and hypertension, drug crystals may further amplify a pre-existing barrier dysfunction and necroinflammation in a crescendo of local intestinal necrosis and systemic inflammation/infection, as occasionally observed in patients on ion-exchange resin therapy.
Collapse
Affiliation(s)
- Tehyung Kim
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, 80336 Munich, Germany; (T.K.); (Q.M.)
| | | | - Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, 80336 Munich, Germany; (T.K.); (Q.M.)
| | - Kathrin Eller
- Division of Nephrology, Department of Internal Medicine, Medical University Graz, 8036 Graz, Austria;
| | - Marion Julia Pollheimer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria;
| | - Danielle Lazarin-Bidóia
- Postgraduate Program in Pharmaceutical Sciences, State University of Maringá, Maringá, Paraná 5790, Brazil; (S.d.O.S.L.); (D.L.-B.); (C.V.N.)
| | - Celso Vataru Nakamura
- Postgraduate Program in Pharmaceutical Sciences, State University of Maringá, Maringá, Paraná 5790, Brazil; (S.d.O.S.L.); (D.L.-B.); (C.V.N.)
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, 80336 Munich, Germany; (T.K.); (Q.M.)
- Correspondence: (H.-J.A.); (S.S.); Tel.: +49-89-4400-53583 (H.-J.A.); +49-89-4400-52129 (S.S.)
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, 80336 Munich, Germany; (T.K.); (Q.M.)
- Correspondence: (H.-J.A.); (S.S.); Tel.: +49-89-4400-53583 (H.-J.A.); +49-89-4400-52129 (S.S.)
| |
Collapse
|
49
|
Rashidi M, Wicks IP, Vince JE. Inflammasomes and Cell Death: Common Pathways in Microparticle Diseases. Trends Mol Med 2020; 26:1003-1020. [DOI: 10.1016/j.molmed.2020.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
|
50
|
Mulay SR, Anders HJ. Neutrophils and Neutrophil Extracellular Traps Regulate Immune Responses in Health and Disease. Cells 2020; 9:E2130. [PMID: 32962213 PMCID: PMC7565859 DOI: 10.3390/cells9092130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Neutrophils are first responders of antimicrobial host defense and sterile inflammation, and therefore, play important roles during health and disease [...].
Collapse
Affiliation(s)
- Shrikant R. Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the LMU Munich, 80336 Munich, Germany
| |
Collapse
|