1
|
Chen Q, Kou H, Demy DL, Liu W, Li J, Wen Z, Herbomel P, Huang Z, Zhang W, Xu J. The different roles of V-ATPase a subunits in phagocytosis/endocytosis and autophagy. Autophagy 2024; 20:2297-2313. [PMID: 38873931 PMCID: PMC11423658 DOI: 10.1080/15548627.2024.2366748] [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: 09/05/2023] [Revised: 04/27/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Microglia are specialized macrophages responsible for the clearance of dead neurons and pathogens by phagocytosis and degradation. The degradation requires phagosome maturation and acidification provided by the vesicular- or vacuolar-type H+-translocating adenosine triphosphatase (V-ATPase), which is composed of the cytoplasmic V1 domain and the membrane-embedded Vo domain. The V-ATPase a subunit, an integral part of the Vo domain, has four isoforms in mammals. The functions of different isoforms on phagosome maturation in different cells/species remain controversial. Here we show that mutations of both the V-ATPase Atp6v0a1 and Tcirg1b/Atp6v0a3 subunits lead to the accumulation of phagosomes in zebrafish microglia. However, their mechanisms are different. The V-ATPase Atp6v0a1 subunit is mainly distributed in early and late phagosomes. Defects of this subunit lead to a defective transition from early phagosomes to late phagosomes. In contrast, The V-ATPase Tcirg1b/Atp6v0a3 subunit is primarily located on lysosomes and regulates late phagosome-lysosomal fusion. Defective Tcirg1b/Atp6v0a3, but not Atp6v0a1 subunit leads to reduced acidification and impaired macroautophagy/autophagy in microglia. We further showed that ATP6V0A1/a1 and TCIRG1/a3 subunits in mouse macrophages preferentially located in endosomes and lysosomes, respectively. Blocking these subunits disrupted early-to-late endosome transition and endosome-to-lysosome fusion, respectively. Taken together, our results highlight the essential and conserved roles played by different V-ATPase subunits in multiple steps of phagocytosis and endocytosis across various species.Abbrevations: Apoe: apolipoprotein E; ANXA5/annexin V: annexin A5; ATP6V0A1/a1: ATPase H+-transporting V0 subunit a1; ATP6V0A2/a2: ATPase H+-transporting V0 subunit a2; ATP6V0A4/a4: ATPase H+-transporting V0 subunit a4; dpf: days post-fertilization; EEA1: early endosome antigen 1; HOPS: homotypic fusion and protein sorting; LAMP1: lysosomal associated membrane protein 1; Lcp1: lymphocyte cytosolic protein 1 (L-plastin); Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; NR: neutral red; PBS: phosphate-buffered saline; PtdIns: phosphatidylinositol; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns(3,5)P2: phosphatidylinositol (3,5)-bisphosphate; RAB4: RAB4, member RAS oncogene family; RAB5: RAB5, member RAS oncogene family; RAB7: RAB7, member RAS oncogene family; TCIRG1/Atp6v0a3/a3: T cell immune regulator 1, ATPase H+-transporting V0 subunit a3; V-ATPase: vacuolar-type H+-translocating adenosine triphosphatase; Xla.Tubb2b/NBT: tubulin beta 2B class IIb.
Collapse
Affiliation(s)
- Qi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PRChina
| | - Hanjing Kou
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Doris Lou Demy
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, France
- CNRS, UMR 3738, Paris, France
| | - Wei Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianchao Li
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Hong Kong, PRChina
| | - Philippe Herbomel
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, France
- CNRS, UMR 3738, Paris, France
| | - Zhibin Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wenqing Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jin Xu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
2
|
Ghorbanzadeh S, Khojini JY, Abouali R, Alimardan S, Zahedi M, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Clearing the Path: Exploring Apoptotic Cell Clearance in Inflammatory and Autoimmune Disorders for Therapeutic Advancements. Mol Biotechnol 2024:10.1007/s12033-024-01222-6. [PMID: 38935260 DOI: 10.1007/s12033-024-01222-6] [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: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 06/28/2024]
Abstract
Inflammatory and autoimmune disorders, characterized by dysregulated immune responses leading to tissue damage and chronic inflammation, present significant health challenges. This review uniquely focuses on efferocytosis-the phagocyte-mediated clearance of apoptotic cells-and its pivotal role in these disorders. We delve into the intricate mechanisms of efferocytosis' four stages and their implications in disease pathogenesis, distinguishing our study from previous literature. Our findings highlight impaired efferocytosis in conditions like atherosclerosis and asthma, proposing its targeting as a novel therapeutic strategy. We discuss the therapeutic potential of efferocytosis in modulating immune responses and resolving inflammation, offering a new perspective in treating inflammatory disorders.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran.
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
3
|
Oliveira AN, Memme JM, Wong J, Hood DA. Dimorphic effect of TFE3 in determining mitochondrial and lysosomal content in muscle following denervation. Skelet Muscle 2024; 14:7. [PMID: 38643162 PMCID: PMC11031958 DOI: 10.1186/s13395-024-00339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/17/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Muscle atrophy is a common consequence of the loss of innervation and is accompanied by mitochondrial dysfunction. Mitophagy is the adaptive process through which damaged mitochondria are removed via the lysosomes, which are regulated in part by the transcription factor TFE3. The role of lysosomes and TFE3 are poorly understood in muscle atrophy, and the effect of biological sex is widely underreported. METHODS Wild-type (WT) mice, along with mice lacking TFE3 (KO), a transcriptional regulator of lysosomal and autophagy-related genes, were subjected to unilateral sciatic nerve denervation for up to 7 days, while the contralateral limb was sham-operated and served as an internal control. A subset of animals was treated with colchicine to capture mitophagy flux. RESULTS WT females exhibited elevated oxygen consumption rates during active respiratory states compared to males, however this was blunted in the absence of TFE3. Females exhibited higher mitophagy flux rates and greater lysosomal content basally compared to males that was independent of TFE3 expression. Following denervation, female mice exhibited less muscle atrophy compared to male counterparts. Intriguingly, this sex-dependent muscle sparing was lost in the absence of TFE3. Denervation resulted in 45% and 27% losses of mitochondrial content in WT and KO males respectively, however females were completely protected against this decline. Decreases in mitochondrial function were more severe in WT females compared to males following denervation, as ROS emission was 2.4-fold higher. In response to denervation, LC3-II mitophagy flux was reduced by 44% in females, likely contributing to the maintenance of mitochondrial content and elevated ROS emission, however this response was dysregulated in the absence of TFE3. While both males and females exhibited increased lysosomal content following denervation, this response was augmented in females in a TFE3-dependent manner. CONCLUSIONS Females have higher lysosomal content and mitophagy flux basally compared to males, likely contributing to the improved mitochondrial phenotype. Denervation-induced mitochondrial adaptations were sexually dimorphic, as females preferentially preserve content at the expense of function, while males display a tendency to maintain mitochondrial function. Our data illustrate that TFE3 is vital for the sex-dependent differences in mitochondrial function, and in determining the denervation-induced atrophy phenotype.
Collapse
Affiliation(s)
- Ashley N Oliveira
- School of Kinesiology and Health Science Muscle Health Research Centre, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Jonathan M Memme
- School of Kinesiology and Health Science Muscle Health Research Centre, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Jenna Wong
- School of Kinesiology and Health Science Muscle Health Research Centre, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - David A Hood
- School of Kinesiology and Health Science Muscle Health Research Centre, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
4
|
Almeida L, Dhillon-LaBrooy A, Sparwasser T. The evolutionary tug-of-war of macrophage metabolism during bacterial infection. Trends Endocrinol Metab 2024; 35:235-248. [PMID: 38040578 DOI: 10.1016/j.tem.2023.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023]
Abstract
The function and phenotype of macrophages are intimately linked with pathogen detection. On sensing pathogen-derived signals and molecules, macrophages undergo a carefully orchestrated process of polarization to acquire pathogen-clearing properties. This phenotypic change must be adequately supported by metabolic reprogramming that is now known to support the acquisition of effector function, but also generates secondary metabolites with direct microbicidal activity. At the same time, bacteria themselves have adapted to both manipulate and take advantage of macrophage-specific metabolic adaptations. Here, we summarize the current knowledge on macrophage metabolism during infection, with a particular focus on understanding the 'arms race' between host immune cells and bacteria during immune responses.
Collapse
Affiliation(s)
- Luís Almeida
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| | - Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| |
Collapse
|
5
|
Zhao M, Chen N, Guo Y, Wu N, Cao B, Zhan B, Li Y, Zhou T, Zhu F, Guo C, Shi Y, Wang Q, Li Y, Zhang L. D-mannose acts as a V-ATPase inhibitor to suppress inflammatory cytokines generation and bacterial killing in macrophage. Mol Immunol 2023; 162:84-94. [PMID: 37660434 DOI: 10.1016/j.molimm.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023]
Abstract
Vacuolar-type H+-ATPase (V-ATPase) critically controls phagosome acidification to promote pathogen digestion and clearance in macrophage. However, the specific subunits of V-ATPase have been evidenced to play contradictory functions in inflammatory cytokines generation and secretion exposure to external bacterial or LPS stimulation. Therefore, identifying the unique function of the separate subunit of V-ATPase is extremely important to regulate macrophage function. Here, we found that D-mannose, a C-2 epimer of glucose, suppressed ATP6V1B2 lysosomal translocation to inhibit V-ATPase activity in macrophages, thereby causing the scaffold protein axis inhibitor protein (AXIN) recruitment to lysosomal membrane and AMPK activation. Correspondingly, LPS-stimulated macrophage M1 polarization was significantly suppressed by D-mannose via down-regulating NF-κB signaling pathway in response to AMPK activation, while IL-4 induced macrophage M2 polarization were not affected. Furthermore, the failure of lysosomal localization of ATP6V1B2 caused by D-mannose also led to the acidification defects of lysosome. Therefore, D-mannose displayed a remarkable function in inhibiting macrophage phagocytosis and bacterial killing. Taken together, D-mannose acts a novel V-ATPase suppressor to attenuate macrophage inflammatory production but simultaneously prevent macrophage phagocytosis and bacterial killing.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Nuo Chen
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Nan Wu
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Baihui Cao
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Bing Zhan
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Shandong University, Jinan, China.
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China.
| |
Collapse
|
6
|
Krishnan V, Nath S, Nair P, Das B. Mycobacterium tuberculosis and its clever approaches to escape the deadly macrophage. World J Microbiol Biotechnol 2023; 39:300. [PMID: 37667129 DOI: 10.1007/s11274-023-03735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/19/2023] [Indexed: 09/06/2023]
Abstract
Mycobacterium tuberculosis (Mt.b), a deadly disease causer, is a facultative parasite. This microorganism has developed several methods to defend itself, once internalized within specialised vacuoles in the macrophages. A wide array of receptors like the complement receptor mannose receptors, scavenger receptor assists the entry of the microbe within the phagocytic macrophages. However, Mt.b is clever enough to protect itself from the hostile environment of the macrophage thereby prevailing within it. The microbe can efficiently inhibit processes like phagosome-lysosome fusion, acidification of phagosomes, release of proinflammatory cytokines and stop crucial events like apoptosis. Additionally, it also adopts resistance to killing by reactive oxygen intermediates and reactive nitrogen intermediates. There are multiple genes both in host and the pathogen which are involved in this successful survival of Mt.b. The regulation of phagolysosome fusion is mediated by proteins such as Coronin, TlyA, SapM, PnkG, EsxH. The microbe has certain mechanisms to even acquire iron from the host cell, to withstand iron deprivation as a mode of host's defence mechanism. This review focuses on the various defensive adaptations acquired by Mt.b for fighting against the deprived conditions existing within the macrophages and their capability of proliferating successfully within it, thereby resulting in a diseased condition.
Collapse
Affiliation(s)
- Vinaya Krishnan
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India
| | | | - Preetha Nair
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India
| | - Bannhi Das
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India.
| |
Collapse
|
7
|
Kim TJ, Shenker BJ, MacElroy AS, Spradlin S, Walker LP, Boesze-Battaglia K. Aggregatibacter actinomycetemcomitans cytolethal distending toxin modulates host phagocytic function. Front Cell Infect Microbiol 2023; 13:1220089. [PMID: 37719670 PMCID: PMC10500838 DOI: 10.3389/fcimb.2023.1220089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023] Open
Abstract
Cytolethal distending toxins (Cdt) are a family of toxins produced by several human pathogens which infect mucocutaneous tissue and induce inflammatory disease. Human macrophages exposed to Aggregatibacter actinomycetemcomitans (Aa) Cdt respond through canonical and non-canonical inflammasome activation to stimulate cytokine release. The inflammatory response is dependent on PI3K signaling blockade via the toxin's phosphatidylinositol-3,4,5-triphosphate (PIP3) phosphatase activity; converting PIP3 to phosphatidylinsoitol-3,4-diphosphate (PI3,4P2) thereby depleting PIP3 pools. Phosphoinositides, also play a critical role in phagosome trafficking, serving as binding domains for effector proteins during phagosome maturation and subsequent fusion with lysosomes. We now demonstrate that AaCdt manipulates the phosphoinositide (PI) pools of phagosome membranes and alters Rab5 association. Exposure of macrophages to AaCdt slowed phagosome maturation and decreased phago-lysosome formation, thereby compromising macrophage phagocytic function. Moreover, macrophages exposed to Cdt showed decreased bactericidal capacity leading to increase in Aggregatibacter actinomycetemcomitans survival. Thus, Cdt may contribute to increased susceptibility to bacterial infection. These studies uncover an underexplored aspect of Cdt function and provide new insight into the virulence potential of Cdt in mediating the pathogenesis of disease caused by Cdt-producing organisms such as Aa.
Collapse
Affiliation(s)
- Taewan J. Kim
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Bruce J. Shenker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Andrew S. MacElroy
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Samuel Spradlin
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lisa P. Walker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
8
|
Mathieu NM, Fekete EM, Muskus PC, Brozoski DT, Lu KT, Wackman KK, Gomez J, Fang S, Reho JJ, Grobe CC, Vazirabad I, Mouradian GC, Hodges MR, Segar JL, Grobe JL, Sigmund CD, Nakagawa P. Genetic Ablation of Prorenin Receptor in the Rostral Ventrolateral Medulla Influences Blood Pressure and Hydromineral Balance in Deoxycorticosterone-Salt Hypertension. FUNCTION 2023; 4:zqad043. [PMID: 37609445 PMCID: PMC10440998 DOI: 10.1093/function/zqad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Non-enzymatic activation of renin via its interaction with prorenin receptor (PRR) has been proposed as a key mechanism of local renin-angiotensin system (RAS) activation. The presence of renin and angiotensinogen has been reported in the rostral ventrolateral medulla (RVLM). Overactivation of bulbospinal neurons in the RVLM is linked to hypertension (HTN). Previous studies have shown that the brain RAS plays a role in the pathogenesis of the deoxycorticosterone (DOCA)-salt HTN model. Thus, we hypothesized that PRR in the RVLM is involved in the local activation of the RAS, facilitating the development of DOCA-salt HTN. Selective PRR ablation targeting the RVLM (PRRRVLM-Null mice) resulted in an unexpected sex-dependent and biphasic phenotype in DOCA-salt HTN. That is, PRRRVLM-Null females (but not males) exhibited a significant delay in achieving maximal pressor responses during the initial stage of DOCA-salt HTN. Female PRRRVLM-Null subsequently showed exacerbated DOCA-salt-induced pressor responses during the "maintenance" phase with a maximal peak at 13 d on DOCA-salt. This exacerbated response was associated with an increased sympathetic drive to the resistance arterioles and the kidney, exacerbated fluid and sodium intake and output in response to DOCA-salt, and induced mobilization of fluids from the intracellular to extracellular space concomitant with elevated vasopressin. Ablation of PRR suppressed genes involved in RAS activation and catecholamine synthesis in the RVLM but also induced expression of genes involved in inflammatory responses. This study illustrates complex and sex-dependent roles of PRR in the neural control of BP and hydromineral balance through autonomic and neuroendocrine systems. Graphical abstract.
Collapse
Affiliation(s)
- Natalia M Mathieu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Eva M Fekete
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Patricia C Muskus
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel T Brozoski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ko-Ting Lu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kelsey K Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Javier Gomez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shi Fang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Connie C Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ibrahim Vazirabad
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gary C Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey L Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Distel JS, Di Venanzio G, Mackel JJ, Rosen DA, Feldman MF. Replicative Acinetobacter baumannii strains interfere with phagosomal maturation by modulating the vacuolar pH. PLoS Pathog 2023; 19:e1011173. [PMID: 37294840 DOI: 10.1371/journal.ppat.1011173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/23/2023] [Indexed: 06/11/2023] Open
Abstract
Bacterial pneumonia is a common infection of the lower respiratory tract that can afflict patients of all ages. Multidrug-resistant strains of Acinetobacter baumannii are increasingly responsible for causing nosocomial pneumonias, thus posing an urgent threat. Alveolar macrophages play a critical role in overcoming respiratory infections caused by this pathogen. Recently, we and others have shown that new clinical isolates of A. baumannii, but not the common lab strain ATCC 19606 (19606), can persist and replicate in macrophages within spacious vacuoles that we called Acinetobacter Containing Vacuoles (ACV). In this work, we demonstrate that the modern A. baumannii clinical isolate 398, but not the lab strain 19606, can infect alveolar macrophages and produce ACVs in vivo in a murine pneumonia model. Both strains initially interact with the alveolar macrophage endocytic pathway, as indicated by EEA1 and LAMP1 markers; however, the fate of these strains diverges at a later stage. While 19606 is eliminated in an autophagy pathway, 398 replicates in ACVs and are not degraded. We show that 398 reverts the natural acidification of the phagosome by secreting large amounts of ammonia, a by-product of amino acid catabolism. We propose that this ability to survive within macrophages may be critical for the persistence of clinical A. baumannii isolates in the lung during a respiratory infection.
Collapse
Affiliation(s)
- Jesus S Distel
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Joseph J Mackel
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - David A Rosen
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Mario F Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
10
|
Filipello F, You SF, Mirfakhar FS, Mahali S, Bollman B, Acquarone M, Korvatska O, Marsh JA, Sivaraman A, Martinez R, Cantoni C, De Feo L, Ghezzi L, Minaya MA, Renganathan A, Cashikar AG, Satoh JI, Beatty W, Iyer AK, Cella M, Raskind WH, Piccio L, Karch CM. Defects in lysosomal function and lipid metabolism in human microglia harboring a TREM2 loss of function mutation. Acta Neuropathol 2023; 145:749-772. [PMID: 37115208 PMCID: PMC10175346 DOI: 10.1007/s00401-023-02568-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
TREM2 is an innate immune receptor expressed by microglia in the adult brain. Genetic variation in the TREM2 gene has been implicated in risk for Alzheimer's disease and frontotemporal dementia, while homozygous TREM2 mutations cause a rare leukodystrophy, Nasu-Hakola disease (NHD). Despite extensive investigation, the role of TREM2 in NHD pathogenesis remains poorly understood. Here, we investigate the mechanisms by which a homozygous stop-gain TREM2 mutation (p.Q33X) contributes to NHD. Induced pluripotent stem cell (iPSC)-derived microglia (iMGLs) were generated from two NHD families: three homozygous TREM2 p.Q33X mutation carriers (termed NHD), two heterozygous mutation carriers, one related non-carrier, and two unrelated non-carriers. Transcriptomic and biochemical analyses revealed that iMGLs from NHD patients exhibited lysosomal dysfunction, downregulation of cholesterol genes, and reduced lipid droplets compared to controls. Also, NHD iMGLs displayed defective activation and HLA antigen presentation. This defective activation and lipid droplet content were restored by enhancing lysosomal biogenesis through mTOR-dependent and independent pathways. Alteration in lysosomal gene expression, such as decreased expression of genes implicated in lysosomal acidification (ATP6AP2) and chaperone mediated autophagy (LAMP2), together with reduction in lipid droplets were also observed in post-mortem brain tissues from NHD patients, thus closely recapitulating in vivo the phenotype observed in iMGLs in vitro. Our study provides the first cellular and molecular evidence that the TREM2 p.Q33X mutation in microglia leads to defects in lysosomal function and that compounds targeting lysosomal biogenesis restore a number of NHD microglial defects. A better understanding of how microglial lipid metabolism and lysosomal machinery are altered in NHD and how these defects impact microglia activation may provide new insights into mechanisms underlying NHD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabia Filipello
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Shih-Feng You
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | | | - Sidhartha Mahali
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Bryan Bollman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Mariana Acquarone
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Olena Korvatska
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Jacob A Marsh
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Anirudh Sivaraman
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Rita Martinez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Claudia Cantoni
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Luca De Feo
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Laura Ghezzi
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Miguel A Minaya
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Arun Renganathan
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Anil G Cashikar
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Jun-Ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Abhirami K Iyer
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Marina Cella
- Department Of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Wendy H Raskind
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Laura Piccio
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA.
- Charles Perkins Centre and Brain and Mind Centre, School of Medical Sciences (Neuroscience), University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Brain and Mind Centre, University of Sydney, 94 Mallett St, Camperdown, Sydney, NSW, 2050, Australia.
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
11
|
Essletzbichler P, Sedlyarov V, Frommelt F, Soulat D, Heinz LX, Stefanovic A, Neumayer B, Superti-Furga G. A genome-wide CRISPR functional survey of the human phagocytosis molecular machinery. Life Sci Alliance 2023; 6:e202201715. [PMID: 36725334 PMCID: PMC9892931 DOI: 10.26508/lsa.202201715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
Phagocytosis, the process by which cells engulf large particles, plays a vital role in driving tissue clearance and host defense. Its dysregulation is connected to autoimmunity, toxic accumulation of proteins, and increased risks for infections. Despite its importance, we lack full understanding of all molecular components involved in the process. To create a functional map in human cells, we performed a genome-wide CRISPRko FACS screen that identified 716 genes. Mapping those hits to a comprehensive protein-protein interaction network annotated for functional cellular processes allowed retrieval of protein complexes identified multiple times and detection of missing phagocytosis regulators. In addition to known components, such as the Arp2/3 complex, the vacuolar-ATPase-Rag machinery, and the Wave-2 complex, we identified and validated new phagocytosis-relevant functions, including the oligosaccharyltransferase complex (MAGT1/SLC58A1, DDOST, STT3B, and RPN2) and the hypusine pathway (eIF5A, DHPS, and DOHH). Overall, our phagocytosis network comprises elements of cargo uptake, shuffling, and biotransformation through the cell, providing a resource for the identification of potential novel drivers for diseases of the endo-lysosomal system. Our approach of integrating protein-protein interaction offers a broadly applicable way to functionally interpret genome-wide screens.
Collapse
Affiliation(s)
- Patrick Essletzbichler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Fabian Frommelt
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Didier Soulat
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Leonhard X Heinz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Adrijana Stefanovic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Benedikt Neumayer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Distel JS, Di Venanzio G, Mackel JJ, Rosen DA, Feldman MF. Replicative Acinetobacter baumannii strains interfere with phagosomal maturation by modulating the vacuolar pH. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526753. [PMID: 36778331 PMCID: PMC9915592 DOI: 10.1101/2023.02.02.526753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Bacterial pneumonia is a common infection of the lower respiratory tract that can afflict patients of all ages. Multidrug-resistant strains of Acinetobacter baumannii are increasingly responsible for causing nosocomial pneumonias, thus posing an urgent threat. Alveolar macrophages play a critical role in overcoming respiratory infections caused by this pathogen. Recently, we and others have shown that new clinical isolates of A. baumannii , but not the common lab strain ATCC 19606 (19606), can persist and replicate in macrophages within spacious vacuoles that we called A cinetobacter C ontaining V acuoles (ACV). In this work, we demonstrate that the modern A. baumannii clinical isolate 398, but not the lab strain 19606, can infect alveolar macrophages and produce ACVs in vivo in a murine pneumonia model. Both strains initially interact with the alveolar macrophage endocytic pathway, as indicated by EEA1 and LAMP1 markers; however, the fate of these strains diverges at a later stage. While 19606 is eliminated in an autophagy pathway, 398 replicates in ACVs and are not degraded. We show that 398 reverts the natural acidification of the phagosome by secreting large amounts of ammonia, a by-product of amino acid catabolism. We propose that this ability to survive within macrophages may be critical for the persistence of clinical A. baumannii isolates in the lung during a respiratory infection.
Collapse
Affiliation(s)
- Jesus S. Distel
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Joseph J. Mackel
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO, United States
| | - David A Rosen
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO, United States
| | - Mario F. Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
13
|
Yu Y, Zhang Z, Walpole GFW, Yu Y. Kinetics of phagosome maturation is coupled to their intracellular motility. Commun Biol 2022; 5:1014. [PMID: 36163370 PMCID: PMC9512794 DOI: 10.1038/s42003-022-03988-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Immune cells degrade internalized pathogens in phagosomes through sequential biochemical changes. The degradation must be fast enough for effective infection control. The presumption is that each phagosome degrades cargos autonomously with a distinct but stochastic kinetic rate. However, here we show that the degradation kinetics of individual phagosomes is not stochastic but coupled to their intracellular motility. By engineering RotSensors that are optically anisotropic, magnetic responsive, and fluorogenic in response to degradation activities in phagosomes, we monitored cargo degradation kinetics in single phagosomes simultaneously with their translational and rotational dynamics. We show that phagosomes that move faster centripetally are more likely to encounter and fuse with lysosomes, thereby acidifying faster and degrading cargos more efficiently. The degradation rates increase nearly linearly with the translational and rotational velocities of phagosomes. Our results indicate that the centripetal motion of phagosomes functions as a clock for controlling the progression of cargo degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Glenn F W Walpole
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA.
| |
Collapse
|
14
|
Baranov MV, Ioannidis M, Balahsioui S, Boersma A, de Boer R, Kumar M, Niwa M, Hirayama T, Zhou Q, Hopkins TM, Grijpstra P, Thutupalli S, Sacanna S, van den Bogaart G. Irregular particle morphology and membrane rupture facilitate ion gradients in the lumen of phagosomes. BIOPHYSICAL REPORTS 2022; 2:100069. [PMID: 36425330 PMCID: PMC9680789 DOI: 10.1016/j.bpr.2022.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/08/2022] [Indexed: 06/16/2023]
Abstract
Localized fluxes, production, and/or degradation coupled to limited diffusion are well known to result in stable spatial concentration gradients of biomolecules in the cell. In this study, we demonstrate that this also holds true for small ions, since we found that the close membrane apposition between the membrane of a phagosome and the surface of the cargo particle it encloses, together with localized membrane rupture, suffice for stable gradients of protons and iron cations within the lumen of the phagosome. Our data show that, in phagosomes containing hexapod-shaped silica colloid particles, the phagosomal membrane is ruptured at the positions of the tips of the rods, but not at other positions. This results in the confined leakage at these positions of protons and iron from the lumen of the phagosome into the cytosol. In contrast, acidification and iron accumulation still occur at the positions of the phagosomes nearer to the cores of the particles. Our study strengthens the concept that coupling metabolic and signaling reaction cascades can be spatially confined by localized limited diffusion.
Collapse
Affiliation(s)
- Maksim V. Baranov
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Melina Ioannidis
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Sami Balahsioui
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Auke Boersma
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Rinse de Boer
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Manoj Kumar
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Masato Niwa
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1–25–4, Daigaku-nishi, Gifu 201–1196, Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1–25–4, Daigaku-nishi, Gifu 201–1196, Japan
| | - Qintian Zhou
- Molecular Design Institute, Department of Chemistry, New York University, New York, NY, United States
| | - Terrence M. Hopkins
- Molecular Design Institute, Department of Chemistry, New York University, New York, NY, United States
| | - Pieter Grijpstra
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Shashi Thutupalli
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- nternational Centre for Theoretical Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Stefano Sacanna
- Molecular Design Institute, Department of Chemistry, New York University, New York, NY, United States
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
- Department of Medical Biology and Pathology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Zhang J, Ding W, Zhao M, Liu J, Xu Y, Wan J, Wang M. Mechanisms of efferocytosis in determining inflammation resolution: Therapeutic potential and the association with cardiovascular disease. Br J Pharmacol 2022; 179:5151-5171. [PMID: 36028471 DOI: 10.1111/bph.15939] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Efferocytosis is defined as the clearance of apoptotic cells (ACs) in physiological and pathological states and is performed by efferocytes, such as macrophages. Efferocytosis can lead to the resolution of inflammation and restore tissue homoeostasis; however, the mechanisms of efferocytosis in determining inflammation resolution are still not completely understood, and the effects of efferocytosis on other proresolving properties need to be explored and explained. In this review, the process of efferocytosis will be summarized briefly, and then these mechanisms and effects will be thoroughly discussed. In addition, the association between the mechanisms of efferocytosis in determining inflammation resolution and cardiovascular diseases will also be reviewed, as an understanding of this association may provide information on novel treatment targets.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China.,department of radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
16
|
Mir MA, Mir B, Kumawat M, Alkhanani M, Jan U. Manipulation and exploitation of host immune system by pathogenic Mycobacterium tuberculosis for its advantage. Future Microbiol 2022; 17:1171-1198. [PMID: 35924958 DOI: 10.2217/fmb-2022-0026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can become a long-term infection by evading the host immune response. Coevolution of Mtb with humans has resulted in its ability to hijack the host's immune systems in a variety of ways. So far, every Mtb defense strategy is essentially dependent on a subtle balance that, if shifted, can promote Mtb proliferation in the host, resulting in disease progression. In this review, the authors summarize many important and previously unknown mechanisms by which Mtb evades the host immune response. Besides recently found strategies by which Mtb manipulates the host molecular regulatory machinery of innate and adaptive immunity, including the intranuclear regulatory machinery, costimulatory molecules, the ubiquitin system and cellular intrinsic immune components will be discussed. A holistic understanding of these immune-evasion mechanisms is of foremost importance for the prevention, diagnosis and treatment of tuberculosis and will lead to new insights into tuberculosis pathogenesis and the development of more effective vaccines and treatment regimens.
Collapse
Affiliation(s)
- Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Bilkees Mir
- Department of Biochemistry & Biochemical Engineering, SHUATS, Allahabad, UP, India
| | - Manoj Kumawat
- Department of Microbiology, Indian Council of Medical Research (ICMR)-NIREH, Bhopal, MP, India
| | - Mustfa Alkhanani
- Biology Department, College of Sciences, University of Hafr Al Batin, P. O. Box 1803, Hafar Al Batin, Saudi Arabia
| | - Ulfat Jan
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| |
Collapse
|
17
|
Hooper KM, Jacquin E, Li T, Goodwin JM, Brumell JH, Durgan J, Florey O. V-ATPase is a universal regulator of LC3-associated phagocytosis and non-canonical autophagy. J Cell Biol 2022; 221:213194. [PMID: 35511089 PMCID: PMC9082624 DOI: 10.1083/jcb.202105112] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 02/04/2022] [Accepted: 04/15/2022] [Indexed: 12/18/2022] Open
Abstract
Non-canonical autophagy is a key cellular pathway in immunity, cancer, and neurodegeneration, characterized by conjugation of ATG8 to endolysosomal single membranes (CASM). CASM is activated by engulfment (endocytosis, phagocytosis), agonists (STING, TRPML1), and infection (influenza), dependent on K490 in the ATG16L1 WD40-domain. However, factors associated with non-canonical ATG16L1 recruitment and CASM induction remain unknown. Here, using pharmacological inhibitors, we investigate a role for V-ATPase during non-canonical autophagy. We report that increased V0–V1 engagement is associated with, and sufficient for, CASM activation. Upon V0–V1 binding, V-ATPase recruits ATG16L1, via K490, during LC3-associated phagocytosis (LAP), STING- and drug-induced CASM, indicating a common mechanism. Furthermore, during LAP, key molecular players, including NADPH oxidase/ROS, converge on V-ATPase. Finally, we show that LAP is sensitive to Salmonella SopF, which disrupts the V-ATPase–ATG16L1 axis and provide evidence that CASM contributes to the Salmonella host response. Together, these data identify V-ATPase as a universal regulator of CASM and indicate that SopF evolved in part to evade non-canonical autophagy.
Collapse
Affiliation(s)
| | - Elise Jacquin
- Signalling Programme, Babraham Institute, Cambridge, UK.,Institut national de la santé et de la recherche médicale UMR-S 1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Taoyingnan Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - John H Brumell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,SickKids Inflammatory Bowel Disease Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joanne Durgan
- Signalling Programme, Babraham Institute, Cambridge, UK
| | - Oliver Florey
- Signalling Programme, Babraham Institute, Cambridge, UK
| |
Collapse
|
18
|
Wang J, Onogi Y, Krueger M, Oeckl J, Karlina R, Singh I, Hauck SM, Feederle R, Li Y, Ussar S. PAT2 regulates vATPase assembly and lysosomal acidification in brown adipocytes. Mol Metab 2022; 61:101508. [PMID: 35513259 PMCID: PMC9114668 DOI: 10.1016/j.molmet.2022.101508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Brown adipocytes play a key role in maintaining body temperature as well as glucose and lipid homeostasis. However, brown adipocytes need to adapt their thermogenic activity and substrate utilization to changes in nutrient availability. Amongst the multiple factors influencing brown adipocyte activity, autophagy is an important regulatory element of thermogenic capacity and activity. Nevertheless, a specific sensing mechanism of extracellular amino acid availability linking autophagy to nutrient availability in brown adipocytes is unknown. METHODS To characterize the role of the amino acid transporter PAT2/SLC36A2 in brown adipocytes, loss or gain of function of PAT2 were studied with respect to differentiation, subcellular localization, lysosomal activity and autophagy. Activity of vATPase was evaluated by quenching of EGFP fused to LC3 or FITC-dextran loaded lysosomes in brown adipocytes upon amino acid starvation, whereas the effect of PAT2 on assembly of the vATPase was investigated by Native-PAGE. RESULTS We show that PAT2 translocates from the plasma membrane to the lysosome in response to amino acid withdrawal. Loss or overexpression of PAT2 impair lysosomal acidification and starvation induced S6K re-phosphorylation, as PAT2 facilitates the assembly of the lysosomal vATPase, by recruitment of the cytoplasmic V1 subunit to the lysosome. CONCLUSION PAT2 is an important sensor of extracellular amino acids and regulator of lysosomal acidification in brown adipocytes.
Collapse
Affiliation(s)
- Jiefu Wang
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Yasuhiro Onogi
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Martin Krueger
- Institute for Anatomy, University of Leipzig, 04103, Leipzig, Germany
| | - Josef Oeckl
- Chair for Molecular Nutritional Medicine TUM School for Life Sciences,Technical University Munich, Munich, Germany
| | - Ruth Karlina
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Inderjeet Singh
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Stefanie M Hauck
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Regina Feederle
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Monoclonal Antibody Core Facility, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine TUM School for Life Sciences,Technical University Munich, Munich, Germany
| | - Siegfried Ussar
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Department of Medicine, Technische Universität München, Munich, Germany.
| |
Collapse
|
19
|
Rudnik S, Heybrock S, Saftig P, Damme M. S-palmitoylation determines TMEM55B-dependent positioning of lysosomes. J Cell Sci 2022; 135:jcs258566. [PMID: 34350967 DOI: 10.1242/jcs.258566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/23/2021] [Indexed: 11/20/2022] Open
Abstract
The spatiotemporal cellular distribution of lysosomes depends on active transport mainly driven by microtubule motors such as kinesins and dynein. Different protein complexes attach these molecular motors to their vesicular cargo. TMEM55B (also known as PIP4P1), as an integral lysosomal membrane protein, is a component of such a complex that mediates the retrograde transport of lysosomes by establishing interactions with the cytosolic scaffold protein JIP4 (also known as SPAG9) and dynein-dynactin. Here, we show that TMEM55B and its paralog TMEM55A (PIP4P2) are S-palmitoylated proteins that are lipidated at multiple cysteine residues. Mutation of all cysteines in TMEM55B prevents S-palmitoylation and causes retention of the mutated protein in the Golgi. Consequently, non-palmitoylated TMEM55B is no longer able to modulate lysosomal positioning and the perinuclear clustering of lysosomes. Additional mutagenesis of the dileucine-based lysosomal sorting motif in non-palmitoylated TMEM55B leads to partial missorting to the plasma membrane instead of retention in the Golgi, implicating a direct effect of S-palmitoylation on the adaptor protein-dependent sorting of TMEM55B. Our data suggest a critical role for S-palmitoylation in the trafficking of TMEM55B and TMEM55B-dependent lysosomal positioning.
Collapse
Affiliation(s)
- Sönke Rudnik
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Saskia Heybrock
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| |
Collapse
|
20
|
Wu Y, Wang MH, Yang T, Qin TY, Qin LL, Hu YM, Zhang CF, Sun BJ, Ding L, Wu LL, Liu TH. Mechanisms for Improving Hepatic Glucolipid Metabolism by Cinnamic Acid and Cinnamic Aldehyde: An Insight Provided by Multi-Omics. Front Nutr 2022; 8:794841. [PMID: 35087857 PMCID: PMC8786797 DOI: 10.3389/fnut.2021.794841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
Cinnamic acid (AC) and cinnamic aldehyde (AL) are two chemicals enriched in cinnamon and have been previously proved to improve glucolipid metabolism, thus ameliorating metabolic disorders. In this study, we employed transcriptomes and proteomes on AC and AL treated db/db mice in order to explore the underlying mechanisms for their effects. Db/db mice were divided into three groups: the control group, AC group and AL group. Gender- and age-matched wt/wt mice were used as a normal group. After 4 weeks of treatments, mice were sacrificed, and liver tissues were used for further analyses. Functional enrichment of differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) were performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. DEPs were further verified by parallel reaction monitoring (PRM). The results suggested that AC and AL share similar mechanisms, and they may improve glucolipid metabolism by improving mitochondrial functions, decreasing serotonin contents and upregulating autophagy mediated lipid clearance. This study provides an insight into the molecular mechanisms of AC and AL on hepatic transcriptomes and proteomes in disrupted metabolic situations and lays a foundation for future experiments.
Collapse
Affiliation(s)
- You Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Ming-Hui Wang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Yang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Tian-Yu Qin
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Ling-Ling Qin
- Department of Science and Technology, Beijing University of Chinese Medicine, Beijing, China
| | - Yao-Mu Hu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng-Fei Zhang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Ju Sun
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Ding
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Li-Li Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Tong-Hua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Sun H, Mao J, Wang Y, Fan Z, Yuan C, Wang X, Tian Y, Han B, Hao Z, Ding J, Chang Y. Quantitative proteomic analysis reveals the molecular mechanism of the Yesso scallop (Patinopecten yessoensis) in response to Polydora infection. Comput Struct Biotechnol J 2022; 20:5966-5977. [PMID: 36382199 PMCID: PMC9641012 DOI: 10.1016/j.csbj.2022.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/08/2022] Open
Abstract
The Yesso scallop is a large and ancient molluscan group with great economic value; however, it has recently suffered severe cases of Polydora infection. Polydora parasitizes the shells of scallops, badly damaging shell structures and affecting growth and mortality. To investigate the molecular mechanism of Yesso scallops’ response to Polydora infection, proteomic profiling changes in the mantle tissues of Polydora-infected (diseased) and healthy scallops were systematically analysed by tandem mass tags (TMT) labelling technology in this study. A total of 519 differentially expressed proteins (DEPs) were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed most innated immune-related functions and pathways were significantly downregulated in diseased scallops, except the phagocytosis pathway, indicating an important role of phagocytosis in response to Polydora infection. DEPs involved in the phagocytosis pathway were associated with phagocytic receptor recognition, phagosome biogenesis and pathogen degradation, and they were further verified by quantitative real-time PCR. The results elucidate the molecular components of phagocytosis in molluscs for the first time. Polydora can be encapsulated by melanization with an obvious appearance in shells; indeed, melanization-related DEPs were upregulated in diseased scallops. Inhibition of apoptosis and nervous modulation may be also involved in the response mechanism, with some highly associated proteins significantly differentially expressed. Finally, a protein–protein interaction network was constructed to provide a global view of the interaction relationships of the DEPs. The study predicts the molecular response mechanism of Yesso scallops to Polydora infection, and lays a theoretical foundation for Polydora disease control.
Collapse
|
22
|
Sang H, Liu J, Zhou F, Zhang X, Zhang J, Liu Y, Wang G, Ye H. Target-responsive subcellular catabolism analysis for early-stage antibody-drug conjugates screening and assessment. Acta Pharm Sin B 2021; 11:4020-4031. [PMID: 35024323 PMCID: PMC8727762 DOI: 10.1016/j.apsb.2021.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Events including antibody‒antigen affinity, internalization, trafficking and lysosomal proteolysis combinatorially determine the efficiency of antibody-drug conjugate (ADC) catabolism and hence the toxicity. Nevertheless, an approach that conveniently identifies proteins requisite for payload release and the ensuing toxicity for mechanistic studies and quality assessment is lacking. Considering the plethora of ADC candidates under development, we developed a target-responsive subcellular catabolism (TARSC) approach that examines ADC catabolism and probes changes in response to targeted interferences of proteins of interest. We firstly applied TARSC to study the commercial T-DM1 and the biosimilar. We recorded unequivocal catabolic behaviors regardless of the absence and presence of the targeted interferences. Their negligible differences in TARSC profiles agreed with their undifferentiated anti-tumoral efficacy according to further in vitro viability and in vivo tumor growth assays, highlighting TARSC analysis as a useful tool for biosimilarity assessment and functional dissection of proteins requisite for ADC catabolism. Additionally, we employed TARSC to investigate the catabolic behavior of a new trastuzumab-toxin conjugate. Collectively, TARSC can not only characterize ADC catabolism at (sub)cellular level but also comprehensively determine which protein targets affect payload release and therapeutic outcomes. Future use of TARSC is thus anticipated in early-stage screening, quality assessment and mechanistic investigations of ADCs.
Collapse
Affiliation(s)
- Hua Sang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacy, the Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jiali Liu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Zhou
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaofang Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwei Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yazhong Liu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors. Tel.: +86 25 83271176 (Guangji Wang), +86 25 83271179 (Hui Ye)
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors. Tel.: +86 25 83271176 (Guangji Wang), +86 25 83271179 (Hui Ye)
| |
Collapse
|
23
|
Abukhalid N, Islam S, Ndzeidze R, Bermudez LE. Mycobacterium avium Subsp. hominissuis Interactions with Macrophage Killing Mechanisms. Pathogens 2021; 10:1365. [PMID: 34832521 PMCID: PMC8623537 DOI: 10.3390/pathogens10111365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Non-tuberculosis mycobacteria (NTM) are ubiquitously found throughout the environment. NTM can cause respiratory infections in individuals with underlying lung conditions when inhaled, or systemic infections when ingested by patients with impaired immune systems. Current therapies can be ineffective at treating NTM respiratory infections, even after a long course or with multidrug treatment regimens. NTM, such as Mycobacterium avium subspecies hominissuis (M. avium), is an opportunistic pathogen that shares environments with ubiquitous free-living amoeba and other environmental hosts, possibly their evolutionary hosts. It is highly likely that interactions between M. avium and free-living amoeba have provided selective pressure on the bacteria to acquire survival mechanisms, which are also used against predation by macrophages. In macrophages, M. avium resides inside phagosomes and has been shown to exit it to infect other cells. M. avium's adaptation to the hostile intra-phagosomal environment is due to many virulence mechanisms. M. avium is able to switch the phenotype of the macrophage to be anti-inflammatory (M2). Here, we have focused on and discussed the bacterial defense mechanisms associated with the intra-phagosome phase of infection. M. avium possesses a plethora of antioxidant enzymes, including the superoxide dismutases, catalase and alkyl hydroperoxide reductase. When these defenses fail or are overtaken by robust oxidative burst, many other enzymes exist to repair damage incurred on M. avium proteins, including thioredoxin/thioredoxin reductase. Finally, M. avium has several oxidant sensors that induce transcription of antioxidant enzymes, oxidation repair enzymes and biofilm- promoting genes. These expressions induce physiological changes that allow M. avium to survive in the face of leukocyte-generated oxidative stress. We will discuss the strategies used by M. avium to infect human macrophages that evolved during its evolution from free-living amoeba. The more insight we gain about M. avium's mode of pathogenicity, the more targets we can have to direct new anti-virulence therapies toward.
Collapse
Affiliation(s)
- Norah Abukhalid
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (N.A.); (S.I.); (R.N.)
| | - Sabrina Islam
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (N.A.); (S.I.); (R.N.)
| | - Robert Ndzeidze
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (N.A.); (S.I.); (R.N.)
| | - Luiz E. Bermudez
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (N.A.); (S.I.); (R.N.)
- Department of Microbiology, College of Science, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
24
|
Hoffmann N, Peters J. Functions of the (pro)renin receptor (Atp6ap2) at molecular and system levels: pathological implications in hypertension, renal and brain development, inflammation, and fibrosis. Pharmacol Res 2021; 173:105922. [PMID: 34607004 DOI: 10.1016/j.phrs.2021.105922] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/16/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
The (pro)renin receptor [(P)RR, Atp6ap2] was initially discovered as a membrane-bound binding partner of prorenin and renin. A soluble (P)RR has additional paracrine effects and is involved in metabolic syndrome and kidney damage. Meanwhile it is clear that most of the effects of the (P)RR are independent of prorenin. In the kidney, (P)RR plays an important role in renal dysfunction by activating proinflammatory and profibrotic molecules. In the brain, (P)RR is expressed in cardiovascular regulatory nuclei and is linked to hypertension. (P)RR is known to be an essential component of the v-ATPase as a key accessory protein and plays an important role in kidney, brain and heart via regulating the pH of the extracellular space and intracellular compartments. V-ATPase and (P)RR together act on WNT and mTOR signalling pathways, which are responsible for cellular homeostasis and autophagy. (P)RR through its role in v-ATPase assembly and function is also important for fast recycling endocytosis by megalin. In the kidney, megalin together with v-ATPase and (P)RR is crucial for endocytic uptake of components of the RAS and their intracellular processing. In the brain, (P)RR, v-ATPases and megalin are important regulators both during development and in the adult. All three proteins are associated with diseases such as XLMR, XMRE, X-linked parkinsonism and epilepsy, cognitive disorders with Parkinsonism, spasticity, intellectual disability, and Alzheimer's Disease which are characterized by impaired neuronal function and/or neuronal loss. The present review focusses on the relevant effects of Atp6ap2 without assigning them necessarily to the RAS. Mechanistically, many effects can be well explained by the role of Atp6ap2 for v-ATPase assembly and function. Furthermore, application of a soluble (P)RR analogue as new therapeutic option is discussed.
Collapse
Affiliation(s)
- Nadin Hoffmann
- Institute of Physiology, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Str. 15A, 17475, Greifswald, Germany
| | - Jörg Peters
- Institute of Physiology, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Str. 15A, 17475, Greifswald, Germany.
| |
Collapse
|
25
|
Chu A, Zirngibl RA, Manolson MF. The V-ATPase a3 Subunit: Structure, Function and Therapeutic Potential of an Essential Biomolecule in Osteoclastic Bone Resorption. Int J Mol Sci 2021; 22:ijms22136934. [PMID: 34203247 PMCID: PMC8269383 DOI: 10.3390/ijms22136934] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
This review focuses on one of the 16 proteins composing the V-ATPase complex responsible for resorbing bone: the a3 subunit. The rationale for focusing on this biomolecule is that mutations in this one protein account for over 50% of osteopetrosis cases, highlighting its critical role in bone physiology. Despite its essential role in bone remodeling and its involvement in bone diseases, little is known about the way in which this subunit is targeted and regulated within osteoclasts. To this end, this review is broadened to include the three other mammalian paralogues (a1, a2 and a4) and the two yeast orthologs (Vph1p and Stv1p). By examining the literature on all of the paralogues/orthologs of the V-ATPase a subunit, we hope to provide insight into the molecular mechanisms and future research directions specific to a3. This review starts with an overview on bone, highlighting the role of V-ATPases in osteoclastic bone resorption. We then cover V-ATPases in other location/functions, highlighting the roles which the four mammalian a subunit paralogues might play in differential targeting and/or regulation. We review the ways in which the energy of ATP hydrolysis is converted into proton translocation, and go in depth into the diverse role of the a subunit, not only in proton translocation but also in lipid binding, cell signaling and human diseases. Finally, the therapeutic implication of targeting a3 specifically for bone diseases and cancer is discussed, with concluding remarks on future directions.
Collapse
|
26
|
Shu L, He Z, Guan X, Yang X, Tian Y, Zhang S, Wu C, He Z, Yan Q, Wang C, Shi Y. A dormant amoeba species can selectively sense and predate on different soil bacteria. Funct Ecol 2021. [DOI: 10.1111/1365-2435.13824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Longfei Shu
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology Sun Yat‐Sen University Guangzhou China
| | - Zhenzhen He
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Xiaotong Guan
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Xueqin Yang
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Yuehui Tian
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Siyi Zhang
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Chenyuan Wu
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Zhili He
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Qingyun Yan
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Cheng Wang
- Environmental Microbiomics Research Center School of Environmental Science and Engineering Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen University Guangzhou China
| | - Yijing Shi
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology Sun Yat‐Sen University Guangzhou China
- School of Environment Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment SCNU Environmental Research InstituteSouth China Normal University Guangzhou China
| |
Collapse
|
27
|
Saxena V, Gao H, Arregui S, Zollman A, Kamocka MM, Xuei X, McGuire P, Hutchens M, Hato T, Hains DS, Schwaderer AL. Kidney intercalated cells are phagocytic and acidify internalized uropathogenic Escherichia coli. Nat Commun 2021; 12:2405. [PMID: 33893305 PMCID: PMC8065053 DOI: 10.1038/s41467-021-22672-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/18/2021] [Indexed: 02/02/2023] Open
Abstract
Kidney intercalated cells are involved in acid-base homeostasis via vacuolar ATPase expression. Here we report six human intercalated cell subtypes, including hybrid principal-intercalated cells identified from single cell transcriptomics. Phagosome maturation is a biological process that increases in biological pathway analysis rank following exposure to uropathogenic Escherichia coli in two of the intercalated cell subtypes. Real time confocal microscopy visualization of murine renal tubules perfused with green fluorescent protein expressing Escherichia coli or pHrodo Green E. coli BioParticles demonstrates that intercalated cells actively phagocytose bacteria then acidify phagolysosomes. Additionally, intercalated cells have increased vacuolar ATPase expression following in vivo experimental UTI. Taken together, intercalated cells exhibit a transcriptional response conducive to the kidney's defense, engulf bacteria and acidify the internalized bacteria. Intercalated cells represent an epithelial cell with characteristics of professional phagocytes like macrophages.
Collapse
Affiliation(s)
- Vijay Saxena
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA.
| | - Hongyu Gao
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Samuel Arregui
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA
| | - Amy Zollman
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - Malgorzata Maria Kamocka
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - Xiaoling Xuei
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Patrick McGuire
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Michael Hutchens
- Oregon Health and Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA
| | - Takashi Hato
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - David S Hains
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA
| | - Andrew L Schwaderer
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA.
| |
Collapse
|
28
|
Hattenrath-Lehmann TK, Nanjappa D, Zhang H, Yu L, Goleski JA, Lin S, Gobler CJ. Transcriptomic and isotopic data reveal central role of ammonium in facilitating the growth of the mixotrophic dinoflagellate, Dinophysis acuminata. HARMFUL ALGAE 2021; 104:102031. [PMID: 34023078 DOI: 10.1016/j.hal.2021.102031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Dinophysis spp. are mixotrophs that are dependent on specific prey, but are also potentially reliant on dissolved nutrients. The extent to which Dinophysis relies on exogenous N and the specific biochemical pathways important for supporting its autotrophic and heterotrophic growth are unknown. Here, the nutritional ecology of Dinophysis was explored using two approaches: 1) 15N tracer experiments were conducted to quantify the concentration-dependent uptake rates and associated kinetics of various N compounds (nitrate, ammonium, urea) of Dinophysis cultures and 2) the transcriptomic responses of Dinophysis cultures grown with multiple combinations of prey and nutrients were assessed via dinoflagellate spliced leader-based transcriptome profiling. Of the N compounds examined, ammonium had the highest Vmax and affinity coefficient, and lowest Ks for both pre-starved and pre-fed cultures, collectively demonstrating the preference of Dinophysis for this N source while little-to-no nitrate uptake was observed. During the transcriptome experiments, Dinophysis grown with nitrate and without prey had the largest number of genes with lower transcript abundances, did not increase abundance of transcripts associated with nitrate/nitrite uptake or reduction, and displayed no cellular growth, suggesting D. acuminata is not capable of growing on nitrate. When offered prey, the transcriptomic response of Dinophysis included the production of phagolysosomes, enzymes involved in protein and lipid catabolism, and N acquisition through amino acid degradation pathways. Compared with cultures only offered ammonium or prey, cultures offered both ammonium and prey had the largest number of genes with increased transcript abundances, the highest growth rate, and the unique activation of multiple pathways involved in cellular catabolism, further evidencing the ability of Dinophysis to grow optimally as a mixotroph. Collectively, this study evidences the key role ammonium plays in the mixotrophic growth of Dinophysis and reveals the precise biochemical pathways that facilitate its mixotrophic growth.
Collapse
Affiliation(s)
- Theresa K Hattenrath-Lehmann
- Stony Brook University, School of Marine and Atmospheric Sciences, 239 Montauk Hwy, Southampton, NY 11968, United States
| | - Deepak Nanjappa
- Stony Brook University, School of Marine and Atmospheric Sciences, 239 Montauk Hwy, Southampton, NY 11968, United States
| | - Huan Zhang
- Department of Marine Sciences, University of Connecticut, Groton, CT 06340, United States
| | - Liying Yu
- State Key Laboratory of Marine Environmental Science and Marine Biodiversity and Global Change Research Center, Xiamen University, Xiamen 361101, China
| | - Jennifer A Goleski
- Stony Brook University, School of Marine and Atmospheric Sciences, 239 Montauk Hwy, Southampton, NY 11968, United States
| | - Senjie Lin
- Department of Marine Sciences, University of Connecticut, Groton, CT 06340, United States; State Key Laboratory of Marine Environmental Science and Marine Biodiversity and Global Change Research Center, Xiamen University, Xiamen 361101, China
| | - Christopher J Gobler
- Stony Brook University, School of Marine and Atmospheric Sciences, 239 Montauk Hwy, Southampton, NY 11968, United States.
| |
Collapse
|
29
|
Nguyen JA, Yates RM. Better Together: Current Insights Into Phagosome-Lysosome Fusion. Front Immunol 2021; 12:636078. [PMID: 33717183 PMCID: PMC7946854 DOI: 10.3389/fimmu.2021.636078] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Following phagocytosis, the nascent phagosome undergoes maturation to become a phagolysosome with an acidic, hydrolytic, and often oxidative lumen that can efficiently kill and digest engulfed microbes, cells, and debris. The fusion of phagosomes with lysosomes is a principal driver of phagosomal maturation and is targeted by several adapted intracellular pathogens. Impairment of this process has significant consequences for microbial infection, tissue inflammation, the onset of adaptive immunity, and disease. Given the importance of phagosome-lysosome fusion to phagocyte function and the many virulence factors that target it, it is unsurprising that multiple molecular pathways have evolved to mediate this essential process. While the full range of these pathways has yet to be fully characterized, several pathways involving proteins such as members of the Rab GTPases, tethering factors and SNAREs have been identified. Here, we summarize the current state of knowledge to clarify the ambiguities in the field and construct a more comprehensive phagolysosome formation model. Lastly, we discuss how other cellular pathways help support phagolysosome biogenesis and, consequently, phagocyte function.
Collapse
Affiliation(s)
- Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Cumming School of Medicine, Snyder Institute of Chronic Disease, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
30
|
Kim JK, Silwal P, Jo EK. Host-Pathogen Dialogues in Autophagy, Apoptosis, and Necrosis during Mycobacterial Infection. Immune Netw 2020; 20:e37. [PMID: 33163245 PMCID: PMC7609165 DOI: 10.4110/in.2020.20.e37] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an etiologic pathogen of human tuberculosis (TB), a serious infectious disease with high morbidity and mortality. In addition, the threat of drug resistance in anti-TB therapy is of global concern. Despite this, it remains urgent to research for understanding the molecular nature of dynamic interactions between host and pathogens during TB infection. While Mtb evasion from phagolysosomal acidification is a well-known virulence mechanism, the molecular events to promote intracellular parasitism remains elusive. To combat intracellular Mtb infection, several defensive processes, including autophagy and apoptosis, are activated. In addition, Mtb-ingested phagocytes trigger inflammation, and undergo necrotic cell death, potentially harmful responses in case of uncontrolled pathological condition. In this review, we focus on Mtb evasion from phagosomal acidification, and Mtb interaction with host autophagy, apoptosis, and necrosis. Elucidation of the molecular dialogue will shed light on Mtb pathogenesis, host defense, and development of new paradigms of therapeutics.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
31
|
Dual Host-Intracellular Parasite Transcriptome of Enucleated Cells Hosting Leishmania amazonensis: Control of Half-Life of Host Cell Transcripts by the Parasite. Infect Immun 2020; 88:IAI.00261-20. [PMID: 32817329 DOI: 10.1128/iai.00261-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
Enucleated cells or cytoplasts (cells whose nucleus is removed in vitro) represent an unexplored biological model for intracellular infection studies due to the abrupt interruption of nuclear processing and new RNA synthesis by the host cell in response to pathogen entry. Using enucleated fibroblasts hosting the protozoan parasite Leishmania amazonensis, we demonstrate that parasite multiplication and biogenesis of large parasitophorous vacuoles in which parasites multiply are independent of the host cell nucleus. Dual RNA sequencing of both host cytoplast and intracellular parasite transcripts identified host transcripts that are more preserved or degraded upon interaction with parasites and also parasite genes that are differentially expressed when hosted by nucleated or enucleated cells. Cytoplasts are suitable host cells, which persist in culture for more than 72 h and display functional enrichment of transcripts related to mitochondrial functions and mRNA translation. Crosstalk between nucleated host de novo gene expression in response to intracellular parasitism and the parasite gene expression to counteract or benefit from these host responses induces a parasite transcriptional profile favoring parasite multiplication and aerobic respiration, and a host-parasite transcriptional landscape enriched in host cell metabolic functions related to NAD, fatty acid, and glycolytic metabolism. Conversely, interruption of host nucleus-parasite cross talk by infection of enucleated cells generates a host-parasite transcriptional landscape in which cytoplast transcripts are enriched in phagolysosome-related pathway, prosurvival, and SerpinB-mediated immunomodulation. In addition, predictive in silico analyses indicated that parasite transcript products secreted within cytoplasts interact with host transcript products conserving the host V-ATPase proton translocation function and glutamine/proline metabolism. The collective evidence indicates parasite-mediated control of host cell transcripts half-life that is beneficial to parasite intracellular multiplication and escape from host immune responses. These findings will contribute to improved drug targeting and serve as database for L. amazonensis-host cell interactions.
Collapse
|
32
|
Aranda-Rivera AK, Cruz-Gregorio A, Briones-Herrera A, Pedraza-Chaverri J. Regulation of autophagy by high- and low-risk human papillomaviruses. Rev Med Virol 2020; 31:e2169. [PMID: 33590566 DOI: 10.1002/rmv.2169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
While high-risk human papillomavirus (HR-HPV) infection is related to the development of cervical, vulvar, anal, penile and oropharyngeal cancer, low-risk human papillomavirus (LR-HPV) infection is implicated in about 90% of genital warts, which rarely progress to cancer. The carcinogenic role of HR-HPV is due to the overexpression of HPV E5, E6 and E7 oncoproteins which target and modify cellular proteins implicated in cell proliferation, apoptosis and immortalization. LR-HPV proteins also target and modify some of these processes; however, their oncogenic potential is lower than that of HR-HPV. HR-HPVs have substantial differences with LR-HPVs such as viral integration into the cell genome, induction of p53 and retinoblastoma protein degradation, alternative splicing in HR-HPV E6-E7 open reading frames, among others. In addition, LR-HPV can activate the autophagy process in infected cells while HR-HPV infection deactivates it. However, in cancer HR-HPV might reactivate autophagy in advance stages. Autophagy is a catabolic process that maintains cell homoeostasis by lysosomal degradation and recycling of damaged macromolecules and organelles; nevertheless, depending upon cellular context autophagy may also induce cell death. Therefore, autophagy can contribute either as a promotor or as a suppressor of tumours. In this review, we focus on the role of HR-HPV and LR-HPV in autophagy during viral infection and cancer development. Additionally, we review key regulatory molecules such as microRNAs in HPV present during autophagy, and we emphasize the potential use of cancer treatments associated with autophagy in HPV-related cancers.
Collapse
Affiliation(s)
- Ana Karina Aranda-Rivera
- Laboratorio 315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, México
| | - Alfredo Cruz-Gregorio
- Laboratorio 225, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| | - Alfredo Briones-Herrera
- Laboratorio 315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, México
| | - José Pedraza-Chaverri
- Laboratorio 315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| |
Collapse
|
33
|
Levin-Konigsberg R, Mantegazza AR. A guide to measuring phagosomal dynamics. FEBS J 2020; 288:1412-1433. [PMID: 32757358 PMCID: PMC7984381 DOI: 10.1111/febs.15506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
Phagocytosis is an essential mechanism for immunity and homeostasis, performed by a subset of cells known as phagocytes. Upon target engulfment, de novo formation of specialized compartments termed phagosomes takes place. Phagosomes then undergo a series of fusion and fission events as they interact with the endolysosomal system and other organelles, in a dynamic process known as phagosome maturation. Because phagocytes play a key role in tissue patrolling and immune surveillance, phagosome maturation is associated with signaling pathways that link phagocytosis to antigen presentation and the development of adaptive immune responses. In addition, and depending on the nature of the cargo, phagosome integrity may be compromised, triggering additional cellular mechanisms including inflammation and autophagy. Upon completion of maturation, phagosomes enter a recently described phase: phagosome resolution, where catabolites from degraded cargo are metabolized, phagosomes are resorbed, and vesicles of phagosomal origin are recycled. Finally, phagocytes return to homeostasis and become ready for a new round of phagocytosis. Altogether, phagosome maturation and resolution encompass a series of dynamic events and organelle crosstalk that can be measured by biochemical, imaging, photoluminescence, cytometric, and immune‐based assays that will be described in this guide.
Collapse
Affiliation(s)
| | - Adriana R Mantegazza
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Bertolini I, Storaci AM, Terrasi A, Cristofori AD, Locatelli M, Caroli M, Ferrero S, Altieri DC, Vaira V. Interplay Between V-ATPase G1 and Small EV-miRNAs Modulates ERK1/2 Activation in GBM Stem Cells and Nonneoplastic Milieu. Mol Cancer Res 2020; 18:1744-1754. [PMID: 32753475 DOI: 10.1158/1541-7786.mcr-20-0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
The ATP6V1G1 subunit (V1G1) of the vacuolar proton ATPase (V-ATPase) pump is crucial for glioma stem cells (GSC) maintenance and in vivo tumorigenicity. Moreover, V-ATPase reprograms the tumor microenvironment through acidification and release of extracellular vesicles (EV). Therefore, we investigated the role of V1G1 in GSC small EVs and their effects on primary brain cultures. To this end, small EVs were isolated from patients-derived GSCs grown as neurospheres (NS) with high (V1G1HIGH-NS) or low (V1G1LOW-NS) V1G1 expression and analyzed for V-ATPase subunits presence, miRNA contents, and cellular responses in recipient cultures. Our results show that NS-derived small EVs stimulate proliferation and motility of recipient cells, with small EV derived from V1G1HIGH-NS showing the most pronounced activity. This involved activation of ERK1/2 signaling, in a response reversed by V-ATPase inhibition in NS-producing small EV. The miRNA profile of V1G1HIGH-NS-derived small EVs differed significantly from that of V1G1LOW-NS, which included miRNAs predicted to target MAPK/ERK signaling. Mechanistically, forced expression of a MAPK-targeting pool of miRNAs in recipient cells suppressed MAPK/ERK pathway activation and blunted the prooncogenic effects of V1G1HIGH small EV. These findings propose that the GSC influences the brain milieu through a V1G1-coordinated EVs release of MAPK/ERK-targeting miRNAs. Interfering with V-ATPase activity could prevent ERK-dependent oncogenic reprogramming of the microenvironment, potentially hampering local GBM infiltration. IMPLICATIONS: Our data identify a novel molecular mechanism of gliomagenesis specific of the GBM stem cell niche, which coordinates a V-ATPase-dependent reprogramming of the brain microenvironment through the release of specialized EVs.
Collapse
Affiliation(s)
- Irene Bertolini
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Maria Storaci
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Terrasi
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Di Cristofori
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Locatelli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Caroli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences University of Milan, Milan, Italy
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
35
|
Huang Y, Zhang Y, Liu Z, Liu C, Zheng J, Qin Q, Huang X. Autophagy Participates in Lysosomal Vacuolation-Mediated Cell Death in RGNNV-Infected Cells. Front Microbiol 2020; 11:790. [PMID: 32425913 PMCID: PMC7212415 DOI: 10.3389/fmicb.2020.00790] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 04/02/2020] [Indexed: 01/14/2023] Open
Abstract
Nervous necrosis virus (NNV) is the etiological agent of viral nervous necrosis (VNN), also known as viral encephalopathy and retinopathy (VER), which results in heavy economic losses to the aquaculture industry worldwide. Dramatic cytoplasmic vacuoles were observed during NNV infection both in vitro and in vivo; however, the origin and mechanism of cytoplasmic vacuolization remains unknown. In this report, we found that the cytoplasmic vacuole morphology became fused and enlarged during infection with red spotted grouper nervous necrosis virus (RGNNV), which was accompanied by increased cell death. Notably, Lyso-Tracker, but not Mito-Tracker or ER-Tracker, was accumulated in the vacuoles, and abnormal lysosome swelling was observed in RGNNV-infected cells, suggesting that the cytoplasmic vacuoles originated from lysosomal organelles. Cytoplasmic vacuolization and cell death in RGNNV-infected cells was completely blocked by the vacuolar H+-ATPase inhibitor (bafilomycin A1), and was significantly weakened by chloroquine (CQ), a lysosomotropic agent that induces the acidification of the lysosomes. This suggests that lysosome acidification was essential for vacuole formation. Significant inhibitory effects on vacuolization and cell death were also observed in the RGNNV-infected cells following treatment with nigericin and monensin (ionophores that uncouple the proton gradient present in lysosomes). This indicated that lysosome function was tightly associated with RGNNV infection-induced cell death. In addition, vacuoles were found to be partially co-localized with GFP-LC3II punctate dots during RGNNV infection. Moreover, the severity of vacuolization and cell death were both significantly decreased after treatment with the autophagy inhibitor, 3-MA, suggesting that autophagy was involved in lysosomal vacuolization and cell death evoked by RGNNV infection. Thus, our results demonstrate that autophagy participates in lysosomal vacuolation-mediated cell death during RGNNV infection, and provides new insight into our understanding of the potential mechanisms underlying nodavirus pathogenesis in vitro.
Collapse
Affiliation(s)
- Youhua Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Ya Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Zetian Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Chuanhe Liu
- Instrumental Analysis & Research Center, South China Agricultural University, Guangzhou, China
| | - Jiaying Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaohong Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
36
|
Boada-Romero E, Martinez J, Heckmann BL, Green DR. The clearance of dead cells by efferocytosis. Nat Rev Mol Cell Biol 2020; 21:398-414. [PMID: 32251387 DOI: 10.1038/s41580-020-0232-1] [Citation(s) in RCA: 407] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
Multiple modes of cell death have been identified, each with a unique function and each induced in a setting-dependent manner. As billions of cells die during mammalian embryogenesis and daily in adult organisms, clearing dead cells and associated cellular debris is important in physiology. In this Review, we present an overview of the phagocytosis of dead and dying cells, a process known as efferocytosis. Efferocytosis is performed by macrophages and to a lesser extent by other 'professional' phagocytes (such as monocytes and dendritic cells) and 'non-professional' phagocytes, such as epithelial cells. Recent discoveries have shed light on this process and how it functions to maintain tissue homeostasis, tissue repair and organismal health. Here, we outline the mechanisms of efferocytosis, from the recognition of dying cells through to phagocytic engulfment and homeostatic resolution, and highlight the pathophysiological consequences that can arise when this process is abrogated.
Collapse
Affiliation(s)
- Emilio Boada-Romero
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer Martinez
- Inflammation & Autoimmunity Group, National Institute for Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
37
|
Lim D, Lee S, Choi M, Han JY, Seong Y, Na D, Kwon Y, Lee YS. The conserved microRNA miR‐8‐3p coordinates the expression of V‐ATPase subunits to regulate ecdysone biosynthesis forDrosophilametamorphosis. FASEB J 2020; 34:6449-6465. [DOI: 10.1096/fj.201901516r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/28/2020] [Accepted: 03/03/2020] [Indexed: 01/17/2023]
Affiliation(s)
- Do‐Hwan Lim
- College of Life Sciences and Biotechnology Korea University Seoul Republic of Korea
- Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| | - Seungjae Lee
- College of Life Sciences and Biotechnology Korea University Seoul Republic of Korea
- Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| | - Min‐Seok Choi
- College of Life Sciences and Biotechnology Korea University Seoul Republic of Korea
- Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| | - Jee Yun Han
- College of Life Sciences and Biotechnology Korea University Seoul Republic of Korea
| | - Youngmo Seong
- Department of Bioscience and Biotechnology Sejong University Seoul Republic of Korea
| | - Dokyun Na
- School of Integrative Engineering Chung‐Ang University Seoul Republic of Korea
| | - Young‐Soo Kwon
- Department of Bioscience and Biotechnology Sejong University Seoul Republic of Korea
| | - Young Sik Lee
- College of Life Sciences and Biotechnology Korea University Seoul Republic of Korea
- Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| |
Collapse
|
38
|
Flinck M, Hagelund S, Gorbatenko A, Severin M, Pedraz-Cuesta E, Novak I, Stock C, Pedersen SF. The Vacuolar H + ATPase α3 Subunit Negatively Regulates Migration and Invasion of Human Pancreatic Ductal Adenocarcinoma Cells. Cells 2020; 9:E465. [PMID: 32085585 PMCID: PMC7072798 DOI: 10.3390/cells9020465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Increased metabolic acid production and upregulation of net acid extrusion render pH homeostasis profoundly dysregulated in many cancers. Plasma membrane activity of vacuolar H+ ATPases (V-ATPases) has been implicated in acid extrusion and invasiveness of some cancers, yet often on the basis of unspecific inhibitors. Serving as a membrane anchor directing V-ATPase localization, the a subunit of the V0 domain of the V-ATPase (ATP6V0a1-4) is particularly interesting in this regard. Here, we map the regulation and roles of ATP6V0a3 in migration, invasion, and growth in pancreatic ductal adenocarcinoma (PDAC) cells. a3 mRNA and protein levels were upregulated in PDAC cell lines compared to non-cancer pancreatic epithelial cells. Under control conditions, a3 localization was mainly endo-/lysosomal, and its knockdown had no detectable effect on pHi regulation after acid loading. V-ATPase inhibition, but not a3 knockdown, increased HIF-1 expression and decreased proliferation and autophagic flux under both starved and non-starved conditions, and spheroid growth of PDAC cells was also unaffected by a3 knockdown. Strikingly, a3 knockdown increased migration and transwell invasion of Panc-1 and BxPC-3 PDAC cells, and increased gelatin degradation in BxPC-3 cells yet decreased it in Panc-1 cells. We conclude that in these PDAC cells, a3 is upregulated and negatively regulates migration and invasion, likely in part via effects on extracellular matrix degradation.
Collapse
Affiliation(s)
- Mette Flinck
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| | - Sofie Hagelund
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| | - Andrej Gorbatenko
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Marc Severin
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| | - Elena Pedraz-Cuesta
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| | - Ivana Novak
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| | - Christian Stock
- Department of Gastroentero-, Hepato- and Endocrinology, Hannover Medical School, D-30625 Hannover, Germany;
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark; (M.F.); (S.H.); (M.S.); (E.P.-C.); (I.N.)
| |
Collapse
|
39
|
de Araujo MEG, Liebscher G, Hess MW, Huber LA. Lysosomal size matters. Traffic 2019; 21:60-75. [PMID: 31808235 PMCID: PMC6972631 DOI: 10.1111/tra.12714] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022]
Abstract
Lysosomes are key cellular catabolic centers that also perform fundamental metabolic, signaling and quality control functions. Lysosomes are not static and they respond dynamically to intra‐ and extracellular stimuli triggering changes in organelle numbers, size and position. Such physical changes have a strong impact on lysosomal activity ultimately influencing cellular homeostasis. In this review, we summarize the current knowledge on lysosomal size regulation, on its physiological role(s) and association to several disease conditions.
Collapse
Affiliation(s)
- Mariana E G de Araujo
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gudrun Liebscher
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.,Austrian Drug Screening Institute, ADSI, Innsbruck, Austria
| |
Collapse
|
40
|
Atp6ap2 deletion causes extensive vacuolation that consumes the insulin content of pancreatic β cells. Proc Natl Acad Sci U S A 2019; 116:19983-19988. [PMID: 31527264 DOI: 10.1073/pnas.1903678116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pancreatic β cells store insulin within secretory granules which undergo exocytosis upon elevation of blood glucose levels. Crinophagy and autophagy are instead responsible to deliver damaged or old granules to acidic lysosomes for intracellular degradation. However, excessive consumption of insulin granules can impair β cell function and cause diabetes. Atp6ap2 is an essential accessory component of the vacuolar ATPase required for lysosomal degradative functions and autophagy. Here, we show that Cre recombinase-mediated conditional deletion of Atp6ap2 in mouse β cells causes a dramatic accumulation of large, multigranular vacuoles in the cytoplasm, with reduction of insulin content and compromised glucose homeostasis. Loss of insulin stores and gigantic vacuoles were also observed in cultured insulinoma INS-1 cells upon CRISPR/Cas9-mediated removal of Atp6ap2. Remarkably, these phenotypic alterations could not be attributed to a deficiency in autophagy or acidification of lysosomes. Together, these data indicate that Atp6ap2 is critical for regulating the stored insulin pool and that a balanced regulation of granule turnover is key to maintaining β cell function and diabetes prevention.
Collapse
|
41
|
Pessoa CC, Reis LC, Ramos-Sanchez EM, Orikaza CM, Cortez C, de Castro Levatti EV, Badaró ACB, Yamamoto JUDS, D’Almeida V, Goto H, Mortara RA, Real F. ATP6V0d2 controls Leishmania parasitophorous vacuole biogenesis via cholesterol homeostasis. PLoS Pathog 2019; 15:e1007834. [PMID: 31199856 PMCID: PMC6594656 DOI: 10.1371/journal.ppat.1007834] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 06/26/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
V-ATPases are part of the membrane components of pathogen-containing vacuoles, although their function in intracellular infection remains elusive. In addition to organelle acidification, V-ATPases are alternatively implicated in membrane fusion and anti-inflammatory functions controlled by ATP6V0d2, the d subunit variant of the V-ATPase complex. Therefore, we evaluated the role of ATP6V0d2 in the biogenesis of pathogen-containing vacuoles using ATP6V0d2 knock-down macrophages infected with the protozoan parasite Leishmania amazonensis. These parasites survive within IFNγ/LPS-activated inflammatory macrophages, multiplying in large/fusogenic parasitophorous vacuoles (PVs) and inducing ATP6V0d2 upregulation. ATP6V0d2 knock-down decreased macrophage cholesterol levels and inhibited PV enlargement without interfering with parasite multiplication. However, parasites required ATP6V0d2 to resist the influx of oxidized low-density lipoprotein (ox-LDL)-derived cholesterol, which restored PV enlargement in ATP6V0d2 knock-down macrophages by replenishing macrophage cholesterol pools. Thus, we reveal parasite-mediated subversion of host V-ATPase function toward cholesterol retention, which is required for establishing an inflammation-resistant intracellular parasite niche.
Collapse
Affiliation(s)
- Carina Carraro Pessoa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Luiza Campos Reis
- Laboratório de Soroepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brasil
| | - Eduardo Milton Ramos-Sanchez
- Laboratório de Soroepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brasil
| | - Cristina Mary Orikaza
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Cristian Cortez
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago de Chile, Chile
| | | | - Ana Carolina Benites Badaró
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | | | - Vânia D’Almeida
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Hiro Goto
- Laboratório de Soroepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brasil
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Fernando Real
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
- * E-mail:
| |
Collapse
|
42
|
Durymanov M, Permyakova A, Sene S, Guo A, Kroll C, Giménez-Marqués M, Serre C, Reineke J. Cellular Uptake, Intracellular Trafficking, and Stability of Biocompatible Metal-Organic Framework (MOF) Particles in Kupffer Cells. Mol Pharm 2019; 16:2315-2325. [DOI: 10.1021/acs.molpharmaceut.8b01185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Mikhail Durymanov
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, 1055 Campanile Avenue, SD-57007 Brookings, South Dakota, United States
- Moscow Institute of Physics and Technology, Institutsky per. 9, 141701, Dolgoprudny, Moscow Region, Russian Federation
| | - Anastasia Permyakova
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, 1055 Campanile Avenue, SD-57007 Brookings, South Dakota, United States
| | - Saad Sene
- Institut des Matériaux Poreux de Paris, FRE 2000 CNRS Ecole Normale Supérieure Ecole Supérieure de Physique et de Chimie Industrielles de Paris, PSL Research University, 75005 Paris, France
| | - Ailin Guo
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, 1055 Campanile Avenue, SD-57007 Brookings, South Dakota, United States
| | - Christian Kroll
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, 1055 Campanile Avenue, SD-57007 Brookings, South Dakota, United States
| | - Mónica Giménez-Marqués
- Institut des Matériaux Poreux de Paris, FRE 2000 CNRS Ecole Normale Supérieure Ecole Supérieure de Physique et de Chimie Industrielles de Paris, PSL Research University, 75005 Paris, France
| | - Christian Serre
- Institut des Matériaux Poreux de Paris, FRE 2000 CNRS Ecole Normale Supérieure Ecole Supérieure de Physique et de Chimie Industrielles de Paris, PSL Research University, 75005 Paris, France
| | - Joshua Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, 1055 Campanile Avenue, SD-57007 Brookings, South Dakota, United States
| |
Collapse
|
43
|
Xia Y, Liu N, Xie X, Bi G, Ba H, Li L, Zhang J, Deng X, Yao Y, Tang Z, Yin B, Wang J, Jiang K, Li Z, Choi Y, Gong F, Cheng X, O'Shea JJ, Chae JJ, Laurence A, Yang XP. The macrophage-specific V-ATPase subunit ATP6V0D2 restricts inflammasome activation and bacterial infection by facilitating autophagosome-lysosome fusion. Autophagy 2019; 15:960-975. [PMID: 30681394 DOI: 10.1080/15548627.2019.1569916] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macroautophagy/autophagy is a conserved ubiquitous pathway that performs diverse roles in health and disease. Although many key, widely expressed proteins that regulate autophagosome formation followed by lysosomal fusion have been identified, the possibilities of cell-specific elements that contribute to the autophagy fusion machinery have not been explored. Here we show that a macrophage-specific isoform of the vacuolar ATPase protein ATP6V0D2/subunit d2 is dispensable for lysosome acidification, but promotes the completion of autophagy via promotion of autophagosome-lysosome fusion through its interaction with STX17 and VAMP8. Atp6v0d2-deficient macrophages have augmented mitochondrial damage, enhanced inflammasome activation and reduced clearance of Salmonella typhimurium. The susceptibility of atp6v0d2 knockout mice to DSS-induced colitis and Salmonella typhimurium-induced death, highlights the in vivo significance of ATP6V0D2-mediated autophagosome-lysosome fusion. Together, our data identify ATP6V0D2 as a key component of macrophage-specific autophagosome-lysosome fusion machinery maintaining macrophage organelle homeostasis and, in turn, limiting both inflammation and bacterial infection. Abbreviations: ACTB/β-actin: actin, beta; ATG14: autophagy related 14; ATG16L1: autophagy related 16-like 1 (S. cerevisiae); ATP6V0D1/2: ATPase, H+ transporting, lysosomal V0 subunit D1/2; AIM2: absent in melanoma 2; BMDM: bone marrow-derived macrophage; CASP1: caspase 1; CGD: chronic granulomatous disease; CSF1/M-CSF: colony stimulating factor 1 (macrophage); CTSB: cathepsin B; DSS: dextran sodium sulfate; IL1B: interleukin 1 beta; IL6: interleukin 6; IRGM: immunity-related GTPase family M member; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1 light chain 3; LPS: lipo-polysaccaride; NLRP3: NLR family, pyrin domain containing 3; PYCARD/ASC: PYD and CARD domain containing; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SNAP29: synaptosomal-associated protein 29; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TLR: toll-like receptor; TNF: tumor necrosis factor ; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1; VAMP8: vesicle-associated membrane protein 8; WT: wild type; 3-MA: 3-methyladenine.
Collapse
Affiliation(s)
- Yu Xia
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Na Liu
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xiuxiu Xie
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Guoyu Bi
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Hongping Ba
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Lin Li
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jinxia Zhang
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xiaofei Deng
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Yao Yao
- b Department of Surgery, Tongji Hospital , Huazhong University of Science and Technology , Wuhan , China
| | - Zhaohui Tang
- b Department of Surgery, Tongji Hospital , Huazhong University of Science and Technology , Wuhan , China
| | - Binjiao Yin
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jing Wang
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Kan Jiang
- c Lymphocyte Cell Biology Section , NIAMS, NIH , Bethesda , MD , USA
| | - Zhuoya Li
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Yongwon Choi
- d Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| | - Feili Gong
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xiang Cheng
- e Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - John J O'Shea
- c Lymphocyte Cell Biology Section , NIAMS, NIH , Bethesda , MD , USA
| | - Jae Jin Chae
- f Inflammatory Disease Section , NHGRI, NIH , Bethesda , MD , USA
| | - Arian Laurence
- g Translational Gastroentology Unit, Nuffield department of medicine, John Radcliffe Hospital , University of Oxford , Oxford , UK
| | - Xiang-Ping Yang
- a Department of Immunology , School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
44
|
Bargen K, Scraba M, Krämer I, Ketterer M, Nehls C, Krokowski S, Repnik U, Wittlich M, Maaser A, Zapka P, Bunge M, Schlesinger M, Huth G, Klees A, Hansen P, Jeschke A, Bendas G, Utermöhlen O, Griffiths G, Gutsmann T, Wohlmann J, Haas A. Virulence‐associated protein A fromRhodococcus equiis an intercompartmental pH‐neutralising virulence factor. Cell Microbiol 2018; 21:e12958. [DOI: 10.1111/cmi.12958] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/17/2018] [Accepted: 09/04/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Kristine Bargen
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Mirella Scraba
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Ina Krämer
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Maren Ketterer
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | | | - Sina Krokowski
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Urska Repnik
- Department of BiosciencesUniversity of Oslo Oslo Norway
| | - Michaela Wittlich
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Anna Maaser
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Pia Zapka
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Madeleine Bunge
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | | | - Gitta Huth
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Annette Klees
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Philipp Hansen
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Andreas Jeschke
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| | - Gerd Bendas
- Pharmaceutical InstituteUniversity of Bonn Bonn Germany
| | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology and Hygiene, University Medical Center, and Center for Molecular Medicine Köln, and German Center for Infection Research (DCIF) Cologne Germany
| | | | | | - Jens Wohlmann
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
- Department of BiosciencesUniversity of Oslo Oslo Norway
| | - Albert Haas
- Division of Biophysics, Cell Biology InstituteUniversity of Bonn Bonn Germany
| |
Collapse
|
45
|
Duan X, Yang S, Zhang L, Yang T. V-ATPases and osteoclasts: ambiguous future of V-ATPases inhibitors in osteoporosis. Theranostics 2018; 8:5379-5399. [PMID: 30555553 PMCID: PMC6276090 DOI: 10.7150/thno.28391] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
Vacuolar ATPases (V-ATPases) play a critical role in regulating extracellular acidification of osteoclasts and bone resorption. The deficiencies of subunit a3 and d2 of V-ATPases result in increased bone density in humans and mice. One of the traditional drug design strategies in treating osteoporosis is the use of subunit a3 inhibitor. Recent findings connect subunits H and G1 with decreased bone density. Given the controversial effects of ATPase subunits on bone density, there is a critical need to review the subunits of V-ATPase in osteoclasts and their functions in regulating osteoclasts and bone remodeling. In this review, we comprehensively address the following areas: information about all V-ATPase subunits and their isoforms; summary of V-ATPase subunits associated with human genetic diseases; V-ATPase subunits and osteopetrosis/osteoporosis; screening of all V-ATPase subunits variants in GEFOS data and in-house data; spectrum of V-ATPase subunits during osteoclastogenesis; direct and indirect roles of subunits of V-ATPases in osteoclasts; V-ATPase-associated signaling pathways in osteoclasts; interactions among V-ATPase subunits in osteoclasts; osteoclast-specific V-ATPase inhibitors; perspective of future inhibitors or activators targeting V-ATPase subunits in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiaohong Duan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Shaoqing Yang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China
| | - Tielin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, People's Republic of China
| |
Collapse
|
46
|
Rao VK, Zavala G, Deb Roy A, Mains RE, Eipper BA. A pH-sensitive luminal His-cluster promotes interaction of PAM with V-ATPase along the secretory and endocytic pathways of peptidergic cells. J Cell Physiol 2018; 234:8683-8697. [PMID: 30317586 DOI: 10.1002/jcp.27528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/11/2018] [Indexed: 01/18/2023]
Abstract
The biosynthetic and endocytic pathways of secretory cells are characterized by progressive luminal acidification, a process which is crucial for posttranslational modifications and membrane trafficking. This progressive fall in luminal pH is mainly achieved by the vacuolar-type-H+ ATPase (V-ATPase). V-ATPases are large, evolutionarily ancient rotary proton pumps that consist of a peripheral V1 complex, which hydrolyzes ATP, and an integral membrane V0 complex, which transports protons from the cytosol into the lumen. Upon sensing the desired luminal pH, V-ATPase activity is regulated by reversible dissociation of the complex into its V1 and V0 components. Molecular details of how intraluminal pH is sensed and transmitted to the cytosol are not fully understood. Peptidylglycine α-amidating mono-oxygenase (PAM; EC 1.14.17.3), a secretory pathway membrane enzyme which shares similar topology with two V-ATPase accessory proteins (Ac45 and prorenin receptor), has a pH-sensitive luminal linker region. Immunofluorescence and sucrose gradient analysis of peptidergic cells (AtT-20) identified distinct subcellular compartments exhibiting spatial co-occurrence of PAM and V-ATPase. In vitro binding assays demonstrated direct binding of the cytosolic domain of PAM to V1H. Blue native PAGE identified heterogeneous high-molecular weight complexes of PAM and V-ATPase. A PAM-1 mutant (PAM-1/H3A) with altered pH sensitivity had diminished ability to form high-molecular weight complexes. In addition, V-ATPase assembly status was altered in PAM-1/H3A expressing cells. Our analysis of the secretory and endocytic pathways of peptidergic cells supports the hypothesis that PAM serves as a luminal pH-sensor, regulating V-ATPase action by altering its assembly status.
Collapse
Affiliation(s)
- Vishwanatha K Rao
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | - Gerardo Zavala
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas
| | - Abhijit Deb Roy
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, Connecticut
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut.,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
47
|
Chakraborty J, Roy S, Murab S, Ravani R, Kaur K, Devi S, Singh D, Sharma S, Mohanty S, Dinda AK, Tandon R, Ghosh S. Modulation of Macrophage Phenotype, Maturation, and Graft Integration through Chondroitin Sulfate Cross-Linking to Decellularized Cornea. ACS Biomater Sci Eng 2018; 5:165-179. [PMID: 33405862 DOI: 10.1021/acsbiomaterials.8b00251] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Decellularized corneas obtained from other species have gained intense popularity in the field of tissue engineering due to its role to serve as an alternative to the limited availability of high-quality donor tissues. However, the decellularized cornea is found to evoke an immune response inspite of the removal of the cellular contents and antigens due to the distortion of the collagen fibrils that exposes certain antigenic sites, which often lead to graft rejection. Therefore, in this study we tested the hypothesis that cross-linking the decellularized corneas with chondroitin sulfate may help in restoring the distorted conformationation changes of fibrous matrix and thus help in reducing the occurrence of graft rejection. Cross-linking of the decellularized cornea with oxidized chondroitin sulfate was validated by ATR-FTIR analysis. An in vitro immune response study involving healthy monocytes and differentiated macrophages with their surface marker analysis by pHrodo red, Lysotracker red, ER tracker, and CD63, LAMP-2 antibodies confirmed that the cross-linked decellularized matrices elicited the least immune response compared to the decellularized ones. We implanted three sets of corneal scaffolds obtained from goat, i.e., native, decellularized, and decellularized corneas conjugated with chondroitin sulfate into the rabbit stroma. Histology analysis, three months after implantation into the rabbit corneal stromal region, confirmed the restoration of the collagen fibril conformation and the migration of cells to the implanted constructs, affirming proper graft integration. Hence we conclude that the chondroitin sulfate cross-linked decellularized corneal matrix may serve as an efficient alternative to the allograft and human cadaveric corneas.
Collapse
Affiliation(s)
- Juhi Chakraborty
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Subhadeep Roy
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sumit Murab
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | | | - Kulwinder Kaur
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | | | | | | | | | | | | | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
48
|
Pamarthy S, Kulshrestha A, Katara GK, Beaman KD. The curious case of vacuolar ATPase: regulation of signaling pathways. Mol Cancer 2018; 17:41. [PMID: 29448933 PMCID: PMC5815226 DOI: 10.1186/s12943-018-0811-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
The Vacuolar ATPase (V-ATPase) is a proton pump responsible for controlling the intracellular and extracellular pH of cells. The structure of V-ATPase has been highly conserved among all eukaryotic cells and is involved in diverse functions across species. V-ATPase is best known for its acidification of endosomes and lysosomes and is also important for luminal acidification of specialized cells. Several reports have suggested the involvement of V-ATPase in maintaining an alkaline intracellular and acidic extracellular pH thereby aiding in proliferation and metastasis of cancer cells respectively. Increased expression of V-ATPase and relocation to the plasma membrane aids in cancer modulates key tumorigenic cell processes like autophagy, Warburg effect, immunomoduation, drug resistance and most importantly cancer cell signaling. In this review, we discuss the direct role of V-ATPase in acidification and indirect regulation of signaling pathways, particularly Notch Signaling.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
49
|
Podinovskaia M, Spang A. The Endosomal Network: Mediators and Regulators of Endosome Maturation. ENDOCYTOSIS AND SIGNALING 2018; 57:1-38. [DOI: 10.1007/978-3-319-96704-2_1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Kissing S, Saftig P, Haas A. Vacuolar ATPase in phago(lyso)some biology. Int J Med Microbiol 2017; 308:58-67. [PMID: 28867521 DOI: 10.1016/j.ijmm.2017.08.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/28/2017] [Accepted: 08/23/2017] [Indexed: 12/23/2022] Open
Abstract
Many eukaryotic cells ingest extracellular particles in a process termed phagocytosis which entails the generation of a new intracellular compartment, the phagosome. Phagosomes change their composition over time and this maturation process culminates in their fusion with acidic, hydrolase-rich lysosomes. During the maturation process, degradation and, when applicable, killing of the cargo may ensue. Many of the events that are pathologically relevant depend on strong acidification of phagosomes by the 'vacuolar' ATPase (V-ATPase). This protein complex acidifies the lumen of some intracellular compartments at the expense of ATP hydrolysis. We discuss here the roles and importance of V-ATPase in intracellular trafficking, its distribution, inhibition and activities, its role in the defense against microorganisms and the counteractivities of pathogens.
Collapse
Affiliation(s)
- Sandra Kissing
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany.
| | - Albert Haas
- Institut für Zellbiologie, Friedrich-Wilhelms-Universität Bonn, Ulrich-Haberland-Str. 61A, D-53121 Bonn, Germany.
| |
Collapse
|