1
|
Ashim J, Seo MJ, Ji S, Heo J, Yu W. Research approaches for exploring the hidden conversations of G protein-coupled receptor transactivation. Mol Pharmacol 2025; 107:100043. [PMID: 40449085 DOI: 10.1016/j.molpha.2025.100043] [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: 12/10/2024] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
G protein-coupled receptor (GPCR) signaling is a crucial physiological mechanism that encompasses a wide range of signaling phenomena. Although traditional GPCR signaling involves G protein or arrestin-related activation, other modes such as biphasic activation, dimer or oligomeric activation, and transactivation have also been observed. Herein, we focus on the increasingly recognized process of GPCR-transactivation. Transactivation refers to the ability of GPCRs to activate other receptor types, especially receptor tyrosine kinases, without engaging their own specific ligands. This cross-talk between GPCRs and other receptors facilitates the integration of multiple signaling pathways, thereby regulating diverse cellular responses, which underscores its physiological significance. In this review, we provide a comprehensive overview of the role of GPCR-transactivation in physiology. We also discuss the growing interest in this field and examine the various tools available for studying transactivation. Additionally, we highlight recent advancements in emerging tools and their application to GPCR-transactivation research. Finally, we propose future research directions and consider the potential impact of new technologies in this rapidly evolving field. SIGNIFICANCE STATEMENT: G protein-coupled receptor transactivation plays a key role in integrating multiple signaling pathways by activating other proteins, like receptor tyrosine kinases, without binding their specific ligands. Here, we focus on the significance of transactivation and the various approaches used to study this phenomenon.
Collapse
Affiliation(s)
- Janbolat Ashim
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Min Jae Seo
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Sangho Ji
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Joongyu Heo
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Wookyung Yu
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea; Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu, Republic of Korea.
| |
Collapse
|
2
|
McGee MY, Enten GA, Boshra SN, Ogunsina O, Gaponenko V, Gao X, Majetschak M. Ethanol promotes protease activated receptor 1: Chemokine (C-X-C motif) receptor 4 heteromerization and enhances thrombin-induced impairment of human lung endothelial cell barrier function. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167335. [PMID: 38969148 PMCID: PMC11330351 DOI: 10.1016/j.bbadis.2024.167335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Ethanol enhances the propensity of PAR1 and CXCR4 to form heteromers. Ethanol increases PAR1:CXCR4 heteromer expression in human lung microvascular endothelial cells (HULEC-5a). Ethanol enhances the efficacy of PAR1 to activate Gα12 upon thrombin stimulation in cells co-expressing CXCR4. Ethanol dose-dependently increases the efficacy of thrombin to impair HULEC-5a barrier function at clinically relevant concentrations. Interference with PAR1:CXCR4 heteromerization mitigates effects of ethanol on thrombin-induced impairment of HULEC-5a barrier function. Our findings provide a molecular mechanism that is likely to contribute to the increased risk of acute respiratory distress syndrome with alcohol abuse.
Collapse
Affiliation(s)
- Michelle Y McGee
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Garrett A Enten
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sadia N Boshra
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Ololade Ogunsina
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, USA
| | - Xianlong Gao
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matthias Majetschak
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
3
|
McGee MY, Ogunsina O, Boshra SN, Gao X, Majetschak M. β-Adrenoceptor Agonists Attenuate Thrombin-Induced Impairment of Human Lung Endothelial Cell Barrier Function and Protect the Lung Vascular Barrier during Resuscitation from Hemorrhagic Shock. Biomedicines 2024; 12:1813. [PMID: 39200278 PMCID: PMC11352179 DOI: 10.3390/biomedicines12081813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
β-adrenoceptor (β-AR) agonists are known to antagonize thrombin-induced impairment (TII) of bovine and ovine lung endothelial barrier function. The effects of adrenoceptor agonists and other vasoactive agents on human lung microvascular endothelial cell (HULEC-5a) barrier function upon thrombin exposure have not been studied. Furthermore, it is unknown whether the in vitro effects of adrenoceptor agonists translate to lung protective effects in vivo. We observed that epinephrine, norepinephrine, and phenylephrine enhanced normal and prevented TII of HULEC-5a barrier function. Arginine vasopressin and angiotensin II were ineffective. α1B-, α2A/B-, and β1/2-ARs were detectable in HULEC-5a by RT-PCR. Propranolol but not doxazosin blocked the effects of all adrenoceptor agonists. Phenylephrine stimulated β2-AR-mediated Gαs activation with 13-fold lower potency than epinephrine. The EC50 to inhibit TII of HULEC-5a barrier function was 1.8 ± 1.9 nM for epinephrine and >100 nM for phenylephrine. After hemorrhagic shock and fluid resuscitation in rats, Evans blue extravasation into the lung increased threefold (p < 0.01 vs. sham). Single low-dose (1.8 μg/kg) epinephrine administration at the beginning of resuscitation had no effects on blood pressure and reduced Evans blue extravasation by 60% (p < 0.05 vs. vehicle). Our findings confirm the effects of β-adrenoceptor agonists in HULEC-5a and suggest that low-dose β-adrenoceptor agonist treatment protects lung vascular barrier function after traumatic hemorrhagic shock.
Collapse
Affiliation(s)
- Michelle Y. McGee
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Ololade Ogunsina
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Sadia N. Boshra
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA
| | - Xianlong Gao
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Matthias Majetschak
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
4
|
Li J, Xi J. Exploring Immune-Related Gene Profiling and Infiltration of Immune Cells in Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma. Genes (Basel) 2024; 15:121. [PMID: 38275602 PMCID: PMC10815177 DOI: 10.3390/genes15010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
Cervical cancer is a widespread malignancy among women, leading to a substantial global health impact. Despite extensive research, our understanding of the basic molecules and pathogenic processes of cervical squamous cell carcinoma is still insufficient. This investigation aims to uncover immune-related genes linked to CESC and delineate their functions. Leveraging data from the GEO and ImmPort databases, a total of 22 immune-related genes were identified. Multiple tools, including DAVID, the human protein atlas, STRING, GeneMANIA, and TCGA, were employed to delve into the expression and roles of these immune genes in CESC, alongside their connections to the disease's pathological features. Through RT-PCR, the study confirmed notable disparities in CXCL8 and CXCL10 mRNA expression between CESC and normal cervical tissue. The TCGA dataset's immune-related information reinforced the association of CXCL8 and CXCL10 with immune infiltration in CESC. This research sheds light on the potential of CXCL8 and CXCL10 as promising therapeutic targets and essential prognostic factors for individuals diagnosed with CESC.
Collapse
Affiliation(s)
- Jialu Li
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou 225012, China;
| | - Juqun Xi
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou 225012, China;
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, China
| |
Collapse
|
5
|
Gao X, Enten GA, McGee MY, Weche M, Majetschak M. α 1-adrenoceptor ligands inhibit chemokine receptor heteromerization partners of α 1B/D-adrenoceptors via interference with heteromer formation. Pharmacol Res 2023; 190:106730. [PMID: 36925091 PMCID: PMC10108735 DOI: 10.1016/j.phrs.2023.106730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/15/2023]
Abstract
We reported previously that α1-adrenoceptor (α1-AR) ligands inhibit chemokine receptor (CR) heteromerization partners of α1B/D-AR. The underlying mechanisms are unknown and in vivo evidence for such effects is missing. Utilizing CCR2 and α1B-AR as prototypical partners, we observed in recombinant systems and THP-1 cells that α1B-AR enhanced whereas its absence inhibited Gαi signaling of CCR2. Phenylephrine and phentolamine reduced the CCR2:α1B-AR heteromerization propensity and inhibited Gαi signaling of CCR2. Phenylephrine cross-recruited β-arrestin-2 to CCR2, and reduced expression of α1B/D-AR, CR partners (CCR1/2, CXCR4) and corresponding heteromers. Phentolamine reduced CR:α1B/D-AR heteromers without affecting β-arrestin-2 recruitment or receptor expression. Phenylephrine/phentolamine prevented leukocyte infiltration mediated via CR heteromerization partners in a murine air pouch model. Our findings document that α1-AR ligands inhibit leukocyte migration mediated by CR heteromerization partners in vivo and suggest interference with α1B-AR:CR heteromerization as a mechanism by which CR partners are inhibited. These findings provide new insights into the pharmacology of GPCR heteromers and indicate that an agonist and antagonist at one GPCR can act as antagonists at heteromerization partners of their target receptors.
Collapse
Affiliation(s)
- Xianlong Gao
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Garrett A Enten
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Michelle Y McGee
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - McWayne Weche
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Matthias Majetschak
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
6
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
7
|
Gao X, DeSantis AJ, Enten GA, Weche M, Marcet JE, Majetschak M. Heteromerization between α 1B -adrenoceptor and chemokine (C-C motif) receptor 2 biases α 1B -adrenoceptor signaling: Implications for vascular function. FEBS Lett 2022; 596:2706-2716. [PMID: 35920096 PMCID: PMC9830583 DOI: 10.1002/1873-3468.14463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 01/12/2023]
Abstract
Previously, we reported that chemokine (C-C motif) receptor 2 (CCR2) heteromerizes with α1B -adrenoceptor (α1B -AR) in leukocytes, through which α1B -AR controls CCR2. Whether such heteromers are expressed in human vascular smooth muscle cells (hVSMCs) is unknown. Bioluminescence resonance energy transfer confirmed formation of recombinant CCR2:α1b -AR heteromers. Proximity ligation assays detected CCR2:α1B -AR heteromers in hVSMCs and human mesenteric arteries. CCR2:α1B -AR heteromerization per se enhanced α1B -AR-mediated Gαq -coupling. Chemokine (C-C motif) ligand 2 (CCL2) binding to CCR2 inhibited Gαq activation via α1B -AR, cross-recruited β-arrestin to and induced internalization of α1B -AR in recombinant systems and in hVSMCs. Our findings suggest that CCR2 within CCR2:α1B -AR heteromers biases α1B -AR signaling and provide a mechanism for previous observations suggesting a role for CCL2/CCR2 in the regulation of cardiovascular function.
Collapse
Affiliation(s)
- Xianlong Gao
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Anthony J DeSantis
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Garrett A Enten
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - McWayne Weche
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jorge E Marcet
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Matthias Majetschak
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
8
|
Yang T, Jiang H, Luo X, Hou Y, Li A, He B, Zhang X, Hao H, Song H, Cai R, Wang X, Wang Y, Yao C, Qi L, Wang Y. Thrombin acts as inducer of proinflammatory macrophage migration inhibitory factor in astrocytes following rat spinal cord injury. J Neuroinflammation 2022; 19:120. [PMID: 35624475 PMCID: PMC9137112 DOI: 10.1186/s12974-022-02488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The danger-associated molecular patterns (DAMPs) are critical contributors to the progressive neuropathology and thereafter affect the functional outcomes following spinal cord injury (SCI). Up to now, the regulatory mechanisms on their inducible production from the living cells remain elusive, aside from their passive release from the necrotic cells. Thrombin is immediately activated by the damaged or stressed central nervous system (CNS), which potently mediates inflammatory astrocytic responses through proteolytic cleavage of protease-activated receptors (PARs). Therefore, SCI-activated thrombin is conceived to induce the production of DAMPs from astrocytes at lesion site. METHODS Rat SCI model was established by the cord contusion at T8-T10. The expression of thrombin and macrophage migration inhibitory factor (MIF) was determined by ELISA and Western blot. The PAR1, PAR3, and PAR4 receptors of thrombin were examined by PCR and immunohistochemistry. Primary astrocytes were isolated and purified from the spinal cord, followed by stimulation with different concentrations of thrombin either for transcriptome sequencing or for analysis of thrombin-mediated expression of MIF and related signal pathways in the presence or absence of various inhibitors. The post-injury locomotor functions were assessed using the Basso, Beattie, and Bresnahan (BBB) locomotor scale. RESULTS MIF protein levels were significantly elevated in parallel with those of thrombin induced by SCI. Immunostaining demonstrated that PAR1 receptor, together with MIF, was abundantly expressed in astrocytes. By transcriptome sequencing and bioinformatical analysis of thrombin-stimulated primary astrocytes, MIF was identified to be dynamically regulated by the serine protease. Investigation of the underlying mechanism using various inhibitors revealed that thrombin-activated PAR1 was responsible for the MIF production of astrocytes through modulation of JNK/NFκB pathway. Administration of PAR1 inhibitor at lesion sites following SCI significantly reduced the protein levels of MIF and ameliorated functional deficits of rat locomotion. CONCLUSION SCI-activated thrombin is a robust inducer of MIF production from astrocytes. Exploring the roles of thrombin in promoting the production of DAMPs from astrocytes at lesion site will provide an alternative strategy for the clinical therapy of CNS inflammation.
Collapse
Affiliation(s)
- Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Haiyan Jiang
- Health Management Center, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Xinye Luo
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Aicheng Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, School of Public Health, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China.
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
9
|
Enten GA, Gao X, Strzelinski HR, Weche M, Liggett SB, Majetschak M. α 1B/D-adrenoceptors regulate chemokine receptor-mediated leukocyte migration via formation of heteromeric receptor complexes. Proc Natl Acad Sci U S A 2022; 119:e2123511119. [PMID: 35537053 PMCID: PMC9171806 DOI: 10.1073/pnas.2123511119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/06/2022] [Indexed: 11/18/2022] Open
Abstract
It is known that catecholamines regulate innate immune functions. The underlying mechanisms, however, are not well understood. Here we show that at least 20 members of the human chemokine receptor (CR) family heteromerize with one or more members of the α1-adrenergic receptor (AR) family in recombinant systems and that such heteromeric complexes are detectable in human monocytes and the monocytic leukemia cell line THP-1. Ligand binding to α1-ARs inhibited migration toward agonists of the CR heteromerization partners of α1B/D-ARs with high potency and 50 to 77% efficacy but did not affect migration induced by a noninteracting CR. Incomplete siRNA knockdown of α1B/D-ARs in THP-1 cells partially inhibited migration toward agonists of their CR heteromerization partners. Complete α1B-AR knockout via CRISPR-Cas9 gene editing in THP-1 cells (THP-1_ADRA1BKO) resulted in 82% reduction of α1D-AR expression and did not affect CR expression. Migration of THP-1_ADRA1BKO cells toward agonists of CR heteromerization partners of α1B/D-ARs was reduced by 82 to 95%. Our findings indicate that CR:α1B/D-AR heteromers are essential for normal function of CR heteromerization partners, provide a mechanism underlying neuroendocrine control of leukocyte trafficking, and offer opportunities to modulate leukocyte and/or cancer cell trafficking in disease processes.
Collapse
Affiliation(s)
- Garrett A. Enten
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Xianlong Gao
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Hannah R. Strzelinski
- Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - McWayne Weche
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Stephen B. Liggett
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
- Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Matthias Majetschak
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| |
Collapse
|
10
|
Plasticity of seven-transmembrane-helix receptor heteromers in human vascular smooth muscle cells. PLoS One 2021; 16:e0253821. [PMID: 34166476 PMCID: PMC8224933 DOI: 10.1371/journal.pone.0253821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/11/2021] [Indexed: 12/24/2022] Open
Abstract
Recently, we reported that the chemokine (C-X-C motif) receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) heteromerize with α1A/B/D-adrenoceptors (ARs) and arginine vasopressin receptor 1A (AVPR1A) in recombinant systems and in rodent and human vascular smooth muscle cells (hVSMCs). In these studies, we observed that heteromerization between two receptor partners may depend on the presence and the expression levels of other partnering receptors. To test this hypothesis and to gain initial insight into the formation of these receptor heteromers in native cells, we utilized proximity ligation assays in hVSMCs to visualize receptor-receptor proximity and systematically studied how manipulation of the expression levels of individual protomers affect heteromerization patterns among other interacting receptor partners. We confirmed subtype-specific heteromerization between endogenously expressed α1A/B/D-ARs and detected that AVPR1A also heteromerizes with α1A/B/D-ARs. siRNA knockdown of CXCR4 and of ACKR3 resulted in a significant re-arrangement of the heteromerization patterns among α1-AR subtypes. Similarly, siRNA knockdown of AVPR1A significantly increased heteromerization signals for seven of the ten receptor pairs between CXCR4, ACKR3, and α1A/B/D-ARs. Our findings suggest plasticity of seven transmembrane helix (7TM) receptor heteromerization in native cells and could be explained by a supramolecular organization of these receptors within dynamic clusters in the plasma membrane. Because we previously observed that recombinant CXCR4, ACKR3, α1a-AR and AVPR1A form hetero-oligomeric complexes composed of 2–4 different protomers, which show signaling properties distinct from individual protomers, re-arrangements of receptor heteromerization patterns in native cells may contribute to the phenomenon of context-dependent GPCR signaling. Furthermore, these findings advise caution in the interpretation of functional consequences after 7TM receptor knockdown in experimental models. Alterations of the heteromerization patterns among other receptor partners may alter physiological and pathological responses, in particular in more complex systems, such as studies on the function of isolated organs or in in vivo experiments.
Collapse
|
11
|
DeSantis AJ, Enten GA, Gao X, Majetschak M. Chemokine receptor antagonists with α 1-adrenergic receptor blocker activity. J Basic Clin Physiol Pharmacol 2021; 33:519-523. [PMID: 34144642 DOI: 10.1515/jbcpp-2020-0523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/14/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Chemokine receptor antagonists are being explored for their therapeutic potential in various disease processes. As the chemokine (C-C motif) receptor 2 (CCR2) antagonist RS504393 is known to compete with ligand binding to α1-adrenoceptors, we tested a panel of 10 CCR antagonists for interactions with α1-adrenoceptors to evaluate potential cardiovascular activities and side-effect profiles. METHODS The PRESTO-Tango β-arrestin recruitment assay was utilized to test whether the CCR antagonists interfere with α1b-AR activation upon stimulation with phenylephrine. Pressure myography with isolated rat resistance arteries was employed to assess their effects on phenylephrine-induced vasoconstriction. The following antagonists were tested: CCR1-BX471, BX513, BI639667; CCR2-RS504393, INCB3284; CCR3-SB328437; and CCR4-AZD2098, and C021; CCR5-Maraviroc; CCR10-BI6901. The pan-α1-adrenoceptor antagonist prazosin was used as control. RESULTS Among the CCR antagonists tested, RS504393, BX513, and C021 inhibited phenylephrine-induced β-arrestin recruitment to α1b-adrenoceptor and phenylephrine-induced vasoconstriction. While RS504393 functioned as a competitive α1-adrenoceptor blocker, BX513 and C021 functioned as noncompetitive α1-adrenoceptor antagonists in both assay systems. Furthermore, RS504393, BX513, and C021 dose-dependently dilated arteries that were fully preconstricted with phenylephrine. CONCLUSIONS Our data suggest that CCR antagonists should be screened for cross-reactivity with α1-adrenoceptors to exclude potential adverse cardiovascular effects when used as anti inflammatory drugs.
Collapse
Affiliation(s)
| | - Garrett A Enten
- Departments of Surgery and Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, USA
| | - Xianlong Gao
- Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Matthias Majetschak
- Departments of Surgery and Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
12
|
Gao X, Enten GA, DeSantis AJ, Majetschak M. Class A G protein-coupled receptors assemble into functional higher-order hetero-oligomers. FEBS Lett 2021; 595:1863-1875. [PMID: 34032285 DOI: 10.1002/1873-3468.14135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022]
Abstract
Although class A seven-transmembrane helix (7TM) receptor hetero-oligomers have been proposed, information on the assembly and function of such higher-order hetero-oligomers is not available. Utilizing bioluminescence resonance energy transfer (BRET), bimolecular luminescence/fluorescence complementation (BiLC/BiFC), and BiLC/BiFC BRET in HEK293T cells, we provide evidence that chemokine (C-X-C motif) receptor 4, atypical chemokine receptor 3, α1a -adrenoceptor, and arginine vasopressin receptor 1A form hetero-oligomers composed of 2-4 different protomers. We show that hetero-oligomerization per se and ligand binding to individual protomers regulate agonist-induced coupling to the signaling transducers of interacting receptor partners. Our findings support the concept that receptor hetero-oligomers form supramolecular machineries with molecular signaling properties distinct from the individual protomers. These findings provide a mechanism for the phenomenon of context-dependent receptor function.
Collapse
Affiliation(s)
- Xianlong Gao
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Garrett A Enten
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Anthony J DeSantis
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Matthias Majetschak
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
13
|
González-Titos A, Hernández-Camarero P, Barungi S, Marchal JA, Kenyon J, Perán M. Trypsinogen and chymotrypsinogen: potent anti-tumor agents. Expert Opin Biol Ther 2021; 21:1609-1621. [PMID: 33896307 DOI: 10.1080/14712598.2021.1922666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Trypsinogen and chymotrypsinogen have been used clinically in tissue repair due to their ability to resolve inflammatory symptoms. Recently, novel evidence has supported the anti-tumourigenic potential of a mixture of trypsinogen and chymotrypsinogen.Areas covered: First, we analyze the structure of these proteases and the effects of pancreatic proteinases on tissue repair, inflammation and the immune system. Second, we summarize studies that provided evidence of the effects of pancreatic (pro)enzymes on tumor cells both in vitro and in vivo and some successful clinical applications of pancreatic (pro)enzymes. Finally, we study pancreatic (pro)enzymes potential molecular targets, such as the proteinase-activated receptors (PARs).Expert opinion: This novel therapy has been shown to have effective antitumor effects. Treatment with these (pro) enzymes sensitizes Cancer Stem Cells (CSCs) which may allow chemotherapy and radiotherapy to be more effective, which could positively affect the recovery of cancer patients.
Collapse
Affiliation(s)
| | | | - Shivan Barungi
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | - Juan Antonio Marchal
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (Ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (Mnat), University of Granada, Granada, Spain
| | - Julian Kenyon
- The Dove Clinic for Integrated Medicine, Twyford, UK
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain.,Excellence Research Unit "Modeling Nature" (Mnat), University of Granada, Granada, Spain
| |
Collapse
|
14
|
Lee C, Viswanathan G, Choi I, Jassal C, Kohlmann T, Rajagopal S. Beta-Arrestins and Receptor Signaling in the Vascular Endothelium. Biomolecules 2020; 11:biom11010009. [PMID: 33374806 PMCID: PMC7824595 DOI: 10.3390/biom11010009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/13/2020] [Accepted: 12/19/2020] [Indexed: 12/17/2022] Open
Abstract
The vascular endothelium is the innermost layer of blood vessels and is a key regulator of vascular tone. Endothelial function is controlled by receptor signaling through G protein-coupled receptors, receptor tyrosine kinases and receptor serine-threonine kinases. The β-arrestins, multifunctional adapter proteins, have the potential to regulate all of these receptor families, although it is unclear as to whether they serve to integrate signaling across all of these different axes. Notably, the β-arrestins have been shown to regulate signaling by a number of receptors important in endothelial function, such as chemokine receptors and receptors for vasoactive substances such as angiotensin II, endothelin-1 and prostaglandins. β-arrestin-mediated signaling pathways have been shown to play central roles in pathways that control vasodilation, cell proliferation, migration, and immune function. At this time, the physiological impact of this signaling has not been studied in detail, but a deeper understanding of it could lead to the development of novel therapies for the treatment of vascular disease.
Collapse
Affiliation(s)
- Claudia Lee
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA;
| | - Gayathri Viswanathan
- Medical Center, Department of Medicine, Division of Cardiology, Duke University, Durham, NC 27710, USA; (G.V.); (I.C.)
| | - Issac Choi
- Medical Center, Department of Medicine, Division of Cardiology, Duke University, Durham, NC 27710, USA; (G.V.); (I.C.)
| | - Chanpreet Jassal
- College of Arts and Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Taylor Kohlmann
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27708, USA;
| | - Sudarshan Rajagopal
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA;
- Medical Center, Department of Medicine, Division of Cardiology, Duke University, Durham, NC 27710, USA; (G.V.); (I.C.)
- Correspondence:
| |
Collapse
|