1
|
Dwyer MB, Aumiller JL, Wedegaertner PB. Going Rogue: Mechanisms, Regulation, and Roles of Mutationally Activated G α in Human Cancer. Mol Pharmacol 2024; 106:198-215. [PMID: 39187387 PMCID: PMC11493338 DOI: 10.1124/molpharm.124.000743] [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: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
G protein-coupled receptors (GPCRs) couple to heterotrimeric G proteins, comprised of α and βγ subunits, to convert extracellular signals into activation of intracellular signaling pathways. Canonically, GPCR-mediated activation results in the exchange of GDP for GTP on G protein α subunits (Gα) and the dissociation of Gα-GTP and G protein βγ subunits (Gβγ), both of which can regulate a variety of signaling pathways. Hydrolysis of bound GTP by Gα returns the protein to Gα-GDP and allows reassociation with Gβγ to reform the inactive heterotrimer. Naturally occurring mutations in Gα have been found at conserved glutamine and arginine amino acids that disrupt the canonical G protein cycle by inhibiting GTP hydrolysis, rendering these mutants constitutively active. Interestingly, these dysregulated Gα mutants are found in many different cancers due to their ability to sustain aberrant signaling without a need for activation by GPCRs. This review will highlight an increased recognition of the prevalence of such constitutively activating Gα mutations in cancers and the signaling pathways activated. In addition, we will discuss new knowledge regarding how these constitutively active Gα are regulated, how different mutations are biochemically distinct, and how mutationally activated Gα are unique compared with GPCR-activated Gα Lastly, we will discuss recent progress in developing inhibitors directly targeting constitutively active Gα mutants. SIGNIFICANCE STATEMENT: Constitutively activating mutations in G protein α subunits (Gα) widely occur in and contribute to the development of many human cancers. To develop ways to inhibit dysregulated, oncogenic signaling by these mutant Gα, it is crucial to better understand mechanisms that lead to constitutive Gα activation and unique mechanisms that regulate mutationally activated Gα in cells. The prevalence of activating mutations in Gα in various cancers makes Gα proteins compelling targets for the development of therapeutics.
Collapse
Affiliation(s)
- Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Wu D, Casey PJ. GPCR-Gα13 Involvement in Mitochondrial Function, Oxidative Stress, and Prostate Cancer. Int J Mol Sci 2024; 25:7162. [PMID: 39000269 PMCID: PMC11241654 DOI: 10.3390/ijms25137162] [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: 04/30/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Gα13 and Gα12, encoded by the GNA13 and GNA12 genes, respectively, are members of the G12 family of Gα proteins that, along with their associated Gβγ subunits, mediate signaling from specific G protein-coupled receptors (GPCRs). Advanced prostate cancers have increased expression of GPCRs such as CXC Motif Chemokine Receptor 4 (CXCR4), lysophosphatidic acid receptor (LPAR), and protease activated receptor 1 (PAR-1). These GPCRs signal through either the G12 family, or through Gα13 exclusively, often in addition to other G proteins. The effect of Gα13 can be distinct from that of Gα12, and the role of Gα13 in prostate cancer initiation and progression is largely unexplored. The oncogenic effect of Gα13 on cell migration and invasion in prostate cancer has been characterized, but little is known about other biological processes such as mitochondrial function and oxidative stress. Current knowledge on the link between Gα13 and oxidative stress is based on animal studies in which GPCR-Gα13 signaling decreased superoxide levels, and the overexpression of constitutively active Gα13 promoted antioxidant gene activation. In human samples, mitochondrial superoxide dismutase 2 (SOD2) correlates with prostate cancer risk and prognostic Gleason grade. However, overexpression of SOD2 in prostate cancer cells yielded conflicting results on cell growth and survival under basal versus oxidative stress conditions. Hence, it is necessary to explore the effect of Gα13 on prostate cancer tumorigenesis, as well as the effect of Gα13 on SOD2 in prostate cancer cell growth under oxidative stress conditions.
Collapse
Affiliation(s)
- Di Wu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore;
| | - Patrick J. Casey
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore;
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, 308 Research Drive, Durham, NC 27710, USA
| |
Collapse
|
3
|
Insights into divalent cation regulation and G 13-coupling of orphan receptor GPR35. Cell Discov 2022; 8:135. [PMID: 36543774 PMCID: PMC9772185 DOI: 10.1038/s41421-022-00499-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/28/2022] [Indexed: 12/24/2022] Open
Abstract
Endogenous ions play important roles in the function and pharmacology of G protein-coupled receptors (GPCRs) with limited atomic evidence. In addition, compared with G protein subtypes Gs, Gi/o, and Gq/11, insufficient structural evidence is accessible to understand the coupling mechanism of G12/13 protein by GPCRs. Orphan receptor GPR35, which is predominantly expressed in the gastrointestinal tract and is closely related to inflammatory bowel diseases (IBDs), stands out as a prototypical receptor for investigating ionic modulation and G13 coupling. Here we report a cryo-electron microscopy structure of G13-coupled GPR35 bound to an anti-allergic drug, lodoxamide. This structure reveals a novel divalent cation coordination site and a unique ionic regulatory mode of GPR35 and also presents a highly positively charged binding pocket and the complementary electrostatic ligand recognition mode, which explain the promiscuity of acidic ligand binding by GPR35. Structural comparison of the GPR35-G13 complex with other G protein subtypes-coupled GPCRs reveals a notable movement of the C-terminus of α5 helix of the Gα13 subunit towards the receptor core and the least outward displacement of the cytoplasmic end of GPR35 TM6. A featured 'methionine pocket' contributes to the G13 coupling by GPR35. Together, our findings provide a structural basis for divalent cation modulation, ligand recognition, and subsequent G13 protein coupling of GPR35 and offer a new opportunity for designing GPR35-targeted drugs for the treatment of IBDs.
Collapse
|
4
|
Olson KM, Campbell A, Alt A, Traynor JR. Finding the Perfect Fit: Conformational Biosensors to Determine the Efficacy of GPCR Ligands. ACS Pharmacol Transl Sci 2022; 5:694-709. [PMID: 36110374 PMCID: PMC9469492 DOI: 10.1021/acsptsci.1c00256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
G protein-coupled receptors (GPCRs) are highly druggable targets that adopt numerous conformations. A ligand's ability to stabilize specific conformation(s) of its cognate receptor determines its efficacy or ability to produce a biological response. Identifying ligands that produce different receptor conformations and potentially discrete pharmacological effects (e.g., biased agonists, partial agonists, antagonists, allosteric modulators) is a major goal in drug discovery and necessary to develop drugs with better effectiveness and fewer side effects. Fortunately, direct measurements of ligand efficacy, via receptor conformational changes are possible with the recent development of conformational biosensors. In this review, we discuss classical efficacy models, including the two-state model, the ternary-complex model, and multistate models. We describe how nanobody-, transducer-, and receptor-based conformational biosensors detect and/or stabilize specific GPCR conformations to identify ligands with different levels of efficacy. In particular, conformational biosensors provide the potential to identify and/or characterize therapeutically desirable but often difficult to measure conformations of receptors faster and better than current methods. For drug discovery/development, several recent proof-of-principle studies have optimized conformational biosensors for high-throughput screening (HTS) platforms. However, their widespread use is limited by the fact that few sensors are reliably capable of detecting low-frequency conformations and technically demanding assay conditions. Nonetheless, conformational biosensors do help identify desirable ligands such as allosteric modulators, biased ligands, or partial agonists in a single assay, representing a distinct advantage over classical methods.
Collapse
Affiliation(s)
- Keith M. Olson
- Department
of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life
Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Andra Campbell
- Department
of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life
Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Andrew Alt
- Department
of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life
Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department
of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United
States
| |
Collapse
|
5
|
Pan Z, Zheng Z, Ye W, Chen C, Ye S. Overexpression of GNA13 correlates with poor prognosis in esophageal squamous cell carcinoma after esophagectomy. Int J Biol Markers 2022; 37:289-295. [PMID: 35706395 DOI: 10.1177/03936155221106799] [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
BACKGROUND This study aimed to explore the expression and clinical implication of guanine nucleotide-binding protein alpha 13 (GNA13) in esophageal squamous cell carcinoma (ESCC). METHODS We first employed western blot analysis to test the GNA13 protein expression level in ESCC tissues. Subsequently, we used immunohistochemistry assays to detect the GNA13 in ESCC specimens from 173 patients who underwent esophagectomy. Survival analysis was performed to define the impact of GNA13 expressions on the prognosis of the ESCC patients based on the clinical and follow-up data. RESULTS The GNA13 protein was shown to be considerably higher in ESCC tissues than in normal esophageal tissues. The level of expression was closely related to the tumor, node, TNM stage, and tumor size. More importantly, ESCC patients with high GNA13 expression carried an increased risk of tumor recurrence compared to those with low GNA13 expression. In addition, a high GNA13 expression level could independently predict worse overall survival and disease-free survival in ESCC. CONCLUSIONS GNA13 could be a novel prognostic biomarker for ESCC patients after esophagectomy.
Collapse
Affiliation(s)
- Zichun Pan
- Department of Oncology, The First Affiliated Hospital, 71068Sun Yat-sen University, Guangzhou, China
| | - Zhousan Zheng
- Department of Oncology, The First Affiliated Hospital, 71068Sun Yat-sen University, Guangzhou, China
| | - Wen Ye
- Department of Oncology, The First Affiliated Hospital, 71068Sun Yat-sen University, Guangzhou, China
| | - Cui Chen
- Department of Oncology, The First Affiliated Hospital, 71068Sun Yat-sen University, Guangzhou, China
| | - Sheng Ye
- Department of Oncology, The First Affiliated Hospital, 71068Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Guo P, Tai Y, Wang M, Sun H, Zhang L, Wei W, Xiang YK, Wang Q. Gα 12 and Gα 13: Versatility in Physiology and Pathology. Front Cell Dev Biol 2022; 10:809425. [PMID: 35237598 PMCID: PMC8883321 DOI: 10.3389/fcell.2022.809425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/17/2022] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs), as the largest family of receptors in the human body, are involved in the pathological mechanisms of many diseases. Heterotrimeric G proteins represent the main molecular switch and receive cell surface signals from activated GPCRs. Growing evidence suggests that Gα12 subfamily (Gα12/13)-mediated signaling plays a crucial role in cellular function and various pathological processes. The current research on the physiological and pathological function of Gα12/13 is constantly expanding, Changes in the expression levels of Gα12/13 have been found in a wide range of human diseases. However, the mechanistic research on Gα12/13 is scattered. This review briefly describes the structural sequences of the Gα12/13 isoforms and introduces the coupling of GPCRs and non-GPCRs to Gα12/13. The effects of Gα12/13 on RhoA and other signaling pathways and their roles in cell proliferation, migration, and immune cell function, are discussed. Finally, we focus on the pathological impacts of Gα12/13 in cancer, inflammation, metabolic diseases, fibrotic diseases, and circulatory disorders are brought to focus.
Collapse
Affiliation(s)
- Paipai Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Manman Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Hanfei Sun
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA, United States.,VA Northern California Health Care System, Mather, CA, United States
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Shields MA, Spaulding C, Metropulos AE, Khalafalla MG, Pham TND, Munshi HG. Gα13 loss in Kras/Tp53 mouse model of pancreatic tumorigenesis promotes tumors susceptible to rapamycin. Cell Rep 2022; 38:110441. [PMID: 35235808 PMCID: PMC8989626 DOI: 10.1016/j.celrep.2022.110441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/22/2021] [Accepted: 02/04/2022] [Indexed: 11/20/2022] Open
Abstract
Gα13 transduces signals from G-protein-coupled receptors. While Gα13 functions as a tumor suppressor in lymphomas, it is not known whether Gα13 is pro-tumorigenic or tumor suppressive in genetically engineered mouse (GEM) models of epithelial cancers. Here, we show that loss of Gα13 in the Kras/Tp53 (KPC) GEM model promotes well-differentiated tumors and reduces survival. Mechanistically, tumors developing in KPC mice with Gα13 loss exhibit increased E-cadherin expression and mTOR signaling. Importantly, human pancreatic ductal adenocarcinoma (PDAC) tumors with low Gα13 expression also exhibit increased E-cadherin expression and mTOR signaling. Treatment with the mTOR inhibitor rapamycin decreases the growth of syngeneic KPC tumors with Gα13 loss by promoting cell death. This work establishes a tumor-suppressive role of Gα13 in pancreatic tumorigenesis in the KPC GEM model and suggests targeting mTOR in human PDAC tumors with Gα13 loss.
Collapse
Affiliation(s)
- Mario A Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA; The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Anastasia E Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA
| | - Mahmoud G Khalafalla
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA
| | - Thao N D Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Lurie 3-220 or Lurie 3-117, Chicago, IL 60611, USA; The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
8
|
Rasheed SAK, Subramanyan LV, Lim WK, Udayappan UK, Wang M, Casey PJ. The emerging roles of Gα12/13 proteins on the hallmarks of cancer in solid tumors. Oncogene 2022; 41:147-158. [PMID: 34689178 PMCID: PMC8732267 DOI: 10.1038/s41388-021-02069-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 01/14/2023]
Abstract
G12 proteins comprise a subfamily of G-alpha subunits of heterotrimeric GTP-binding proteins (G proteins) that link specific cell surface G protein-coupled receptors (GPCRs) to downstream signaling molecules and play important roles in human physiology. The G12 subfamily contains two family members: Gα12 and Gα13 (encoded by the GNA12 and GNA13 genes, respectively) and, as with all G proteins, their activity is regulated by their ability to bind to guanine nucleotides. Increased expression of both Gα12 and Gα13, and their enhanced signaling, has been associated with tumorigenesis and tumor progression of multiple cancer types over the past decade. Despite these strong associations, Gα12/13 proteins are underappreciated in the field of cancer. As our understanding of G protein involvement in oncogenic signaling has evolved, it has become clear that Gα12/13 signaling is pleotropic and activates specific downstream effectors in different tumor types. Further, the expression of Gα12/13 proteins is regulated through a series of transcriptional and post-transcriptional mechanisms, several of which are frequently deregulated in cancer. With the ever-increasing understanding of tumorigenic processes driven by Gα12/13 proteins, it is becoming clear that targeting Gα12/13 signaling in a context-specific manner could provide a new strategy to improve therapeutic outcomes in a number of solid tumors. In this review, we detail how Gα12/13 proteins, which were first discovered as proto-oncogenes, are now known to drive several "classical" hallmarks, and also play important roles in the "emerging" hallmarks, of cancer.
Collapse
Affiliation(s)
| | | | - Wei Kiang Lim
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Udhaya Kumari Udayappan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Patrick J Casey
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Dept. of Pharmacology and Cancer Biology, Duke Univ. Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
9
|
Bettegazzi B, Bellani S, Cattaneo S, Codazzi F, Grohovaz F, Zacchetti D. Gα13 Contributes to LPS-Induced Morphological Alterations and Affects Migration of Microglia. Mol Neurobiol 2021; 58:6397-6414. [PMID: 34529232 DOI: 10.1007/s12035-021-02553-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022]
Abstract
Microglia are the resident immune cells of the CNS that are activated in response to a variety of stimuli. This phenotypical change is aimed to maintain the local homeostasis, also by containing the insults and repair the damages. All these processes are tightly regulated and coordinated and a failure in restoring homeostasis by microglia can result in the development of neuroinflammation that can facilitate the progression of pathological conditions. Indeed, chronic microglia activation is commonly recognized as a hallmark of many neurological disorders, especially at an early stage. Many complex pathways, including cytoskeletal remodeling, are involved in the control of the microglial phenotypical and morphological changes that occur during activation. In this work, we focused on the small GTPase Gα13 and its role at the crossroad between RhoA and Rac1 signaling when microglia is exposed to pro-inflammatory stimulation. We propose the direct involvement of Gα13 in the cytoskeletal rearrangements mediated by FAK, LIMK/cofilin, and Rac1 during microglia activation. In fact, we show that Gα13 knockdown significantly inhibited LPS-induced microglial cell activation, in terms of both changes in morphology and migration, through the modulation of FAK and one of its downstream effectors, Rac1. In conclusion, we propose Gα13 as a critical factor in the regulation of morphological and functional properties of microglia during activation, which might become a target of intervention for the control of microglia inflammation.
Collapse
Affiliation(s)
- Barbara Bettegazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy.
| | - Serena Bellani
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Stefano Cattaneo
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Franca Codazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Fabio Grohovaz
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Daniele Zacchetti
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
10
|
Olson KM, Traynor JR, Alt A. Allosteric Modulator Leads Hiding in Plain Site: Developing Peptide and Peptidomimetics as GPCR Allosteric Modulators. Front Chem 2021; 9:671483. [PMID: 34692635 PMCID: PMC8529114 DOI: 10.3389/fchem.2021.671483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Allosteric modulators (AMs) of G-protein coupled receptors (GPCRs) are desirable drug targets because they can produce fewer on-target side effects, improved selectivity, and better biological specificity (e.g., biased signaling or probe dependence) than orthosteric drugs. An underappreciated source for identifying AM leads are peptides and proteins-many of which were evolutionarily selected as AMs-derived from endogenous protein-protein interactions (e.g., transducer/accessory proteins), intramolecular receptor contacts (e.g., pepducins or extracellular domains), endogenous peptides, and exogenous libraries (e.g., nanobodies or conotoxins). Peptides offer distinct advantages over small molecules, including high affinity, good tolerability, and good bioactivity, and specific disadvantages, including relatively poor metabolic stability and bioavailability. Peptidomimetics are molecules that combine the advantages of both peptides and small molecules by mimicking the peptide's chemical features responsible for bioactivity while improving its druggability. This review 1) discusses sources and strategies to identify peptide/peptidomimetic AMs, 2) overviews strategies to convert a peptide lead into more drug-like "peptidomimetic," and 3) critically analyzes the advantages, disadvantages, and future directions of peptidomimetic AMs. While small molecules will and should play a vital role in AM drug discovery, peptidomimetics can complement and even exceed the advantages of small molecules, depending on the target, site, lead, and associated factors.
Collapse
Affiliation(s)
- Keith M. Olson
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
| | - John R. Traynor
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Andrew Alt
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Li Y, Long X, Wang J, Peng J, Shen K. miRNA-128 modulates bone neoplasms cells proliferation and migration through the WNT/β-catenin and EMT signal pathways. J Orthop Surg Res 2021; 16:71. [PMID: 33472642 PMCID: PMC7816476 DOI: 10.1186/s13018-020-02164-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/17/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Bone neoplasms present poor prognosis due to recurrence and metastasis. Although the role microRNAs (miRNAs) in inhibiting growth and metastasis of bone neoplasms has been investigated, the underlying potential molecular mechanisms mediated by miRNA-128 (miR-218) for the invasiveness of bone neoplasms cells are still not completely understood. The purpose of this study was to identify the regulatory mechanisms of miR-218 in bone neoplasms cells. METHODS Western blotting, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Counting Kit-8 assay, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, luciferase activity assay immunofluorescence, and immunohistochemistry were used to analyze the regulatory effects of miR-218 on bone neoplasms cells. RESULTS Here, the results showed that transfection of miR-128 suppressed bone neoplasms cells proliferation, migration, and invasion. Genetic knockdown of miR-128 in bone neoplasms cells suppressed the activation of the Wnt/β-catenin and epithelial-mesenchymal transition (EMT) signaling pathways. Activation of Wnt or EMT blocked miR-128-inhibited cells proliferation and migration in bone neoplasms cells. Exogenously introduced miR-128 markedly inhibited tumor regeneration in bone neoplasms xenograft models. CONCLUSIONS These results define a tumor-regulated function for miR-128 in bone neoplasms by down-regulation of the Wnt/β-catenin and EMT signal pathways, which provided a potential target for bone neoplasms gene therapy.
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopedics, Chongqing General Hospital, University of the Chinese Academy of Sciences, No. 312 Zhongshanyi Road, Yuzhong District, Chongqing, 400013, China
| | - Xiaotao Long
- Department of Orthopedics, Chongqing General Hospital, University of the Chinese Academy of Sciences, No. 312 Zhongshanyi Road, Yuzhong District, Chongqing, 400013, China
| | - Ji Wang
- Department of Orthopedics, Chongqing General Hospital, University of the Chinese Academy of Sciences, No. 312 Zhongshanyi Road, Yuzhong District, Chongqing, 400013, China
| | - Jing Peng
- Department of Orthopedics, Chongqing General Hospital, University of the Chinese Academy of Sciences, No. 312 Zhongshanyi Road, Yuzhong District, Chongqing, 400013, China
| | - Kai Shen
- Department of Orthopedics, Chongqing General Hospital, University of the Chinese Academy of Sciences, No. 312 Zhongshanyi Road, Yuzhong District, Chongqing, 400013, China.
| |
Collapse
|