1
|
He LM, Borjigin S, Chen XQ, Yan ZL, Wang MJ. Therapeutic potential of integrins in diabetic retinopathy. World J Diabetes 2025; 16:101509. [DOI: 10.4239/wjd.v16.i5.101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/17/2025] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
Diabetic retinopathy (DR), a leading cause of visual loss, is the result of microvascular damage induced by prolonged hyperglycemia. Numerous studies have revealed the pivotal role of integrins in the pathogenesis of DR, particularly in key processes such as inflammation, vascular leakage, microthrombus formation, and angiogenesis. Consequently, targeting integrins is considered a promising strategy for the treatment of DR. This review focuses on the function of integrins in DR and their potential as therapeutic targets. It describes the molecular mechanisms through which integrins influence DR progression and summarizes the latest outcomes of integrin antagonist-based therapeutic strategies in clinical studies, evaluating their efficacy and potential challenges, which offer promise as novel treatment options for DR.
Collapse
Affiliation(s)
- Li-Mei He
- Department of Internal Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Sarul Borjigin
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Xin-Qi Chen
- Department of Internal Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Zhao-Li Yan
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Ming-Jie Wang
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
2
|
Chen YC, Huang CH, Chang YT, Chen CY, Shiu JH, Cheng CH, Su YF, Chuang WJ. Structural and Functional Differences of Rhodostomin and Echistatin in Integrin Recognition and Biological Implications. Proteins 2025. [PMID: 40318183 DOI: 10.1002/prot.26834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
Rhodostomin (Rho) and Echistatin (Ech) are RGD-containing disintegrins with different sizes, disulfide bond patterns, and amino acid sequences in their RGD loops and C-termini. Cell adhesion analyzes showed that Rho exhibited a 5.2-, 18.9-, 2.2-, and 1.7-fold lower inhibitory activity against integrins αvβ3, α5β1, αIIbβ3, and αvβ5 in comparison with those of Ech. In contrast, Rho exhibited an 8.8-fold higher activity than Ech in inhibiting integrin αvβ6. The swapping of Ech's RGD loop and C-terminal sequences into those of Rho cannot increase its integrins' inhibitory activities. Interestingly, the mutation of Ech into Rho's RGD loop PRGDMP sequence and C-terminal YH sequence caused an 8.2-fold higher activity in inhibiting integrin αvβ6. Structural analyzes of Rho and Ech showed that they have similar conformations in their RGD loop and different conformations in their C-terminal regions. Molecular docking found that not only the RGD loop but also the C-terminal region of Rho and Ech interacted with integrins, showing that the C-terminal region is also important for integrin recognition. The docking of Rho into integrin αvβ6 showed that the C-terminal H68 residue of Rho interacted with D129 of β6. In contrast, the docking of Ech into integrin α5β1 showed that the C-terminal H44 residue of Ech interacted with Q191 of β1. Ech exhibited 78.5- and 10.9-fold higher activities in inhibiting HUVEC proliferation and A375 melanoma cell migration than those of Rho. These findings demonstrate that the disulfide bond pattern, RGD loop, and C-terminal region of disintegrins may cause their functional differences. The functional and structural differences between Rho and Ech support their potential as scaffolds to design drugs targeting their respective integrins.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chun-Hao Huang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Yao-Tsung Chang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chiu-Yueh Chen
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Jia-Hau Shiu
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chun-Ho Cheng
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Yu-Fang Su
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Woei-Jer Chuang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| |
Collapse
|
3
|
Lemay SE, Montesinos MS, Grobs Y, Yokokawa T, Shimauchi T, Mougin M, Romanet C, Sauvaget M, Breuils-Bonnet S, Bourgeois A, Théberge C, Pelletier A, El Kabbout R, Martineau S, Yamamoto K, Akram M, Ray AS, Lippa B, Goodwin B, Lin FY, Wang H, Dowling JE, Lu M, Qiao Q, McTeague TA, Moy TI, Potus F, Provencher S, Boucherat O, Bonnet S. Exploring Integrin α5β1 as a Potential Therapeutic Target for Pulmonary Arterial Hypertension: Insights From Comprehensive Multicenter Preclinical Studies. Circulation 2025; 151:1162-1183. [PMID: 39829438 PMCID: PMC12011439 DOI: 10.1161/circulationaha.124.070693] [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: 05/27/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by obliterative vascular remodeling of the small pulmonary arteries (PAs) and progressive increase in pulmonary vascular resistance leading to right ventricular failure. Although several drugs are approved for the treatment of PAH, mortality rates remain high. Accumulating evidence supports a pathological function of integrins in vessel remodeling, which are gaining renewed interest as drug targets. However, their role in PAH remains largely unexplored. METHODS The expression of the RGD (arginylglycylaspartic acid)-binding integrin α5β1 was assessed in PAs, PA smooth muscle cells, and PA endothelial cells from patients with PAH and controls using NanoString, immunoblotting, and Mesoscale Discovery assays. RNA sequencing was conducted to identify gene networks regulated by α5β1 inhibition in PAH PA smooth muscle cells. The therapeutic efficacy of α5β1 inhibition was evaluated using a novel small molecule inhibitor and selective neutralizing antibodies in Sugen/hypoxia and monocrotaline rat models, with validation by an external contract research organization. Comparisons were made against standard-of-care therapies (ie, macitentan, tadalafil) and sotatercept and efficacy was assessed using echocardiographic, hemodynamic, and histological assessments. Ex vivo studies using human precision-cut lung slices were performed to further assess the effects of α5β1 inhibition on pulmonary vascular remodeling. RESULTS We found that the arginine-glycine-aspartate RGD-binding integrin α5β1 is upregulated in PA endothelial cells and PA smooth muscle cells from patients with PAH and remodeled PAs from animal models. Blockade of the integrin α5β1 or depletion of the α5 subunit downregulated FOXM1 (forkhead box protein M1)-regulated gene networks, resulting in mitotic defects and inhibition of the pro-proliferative and apoptosis-resistant phenotype of PAH cells. We demonstrated that α5β1 integrin blockade safely attenuates pulmonary vascular remodeling and improves hemodynamics and right ventricular function and matched or exceeded the efficacy of standard of care and sotatercept in multiple preclinical models. Ex vivo studies further validated its potential in reversing advanced remodeling in human precision-cut lung slices. CONCLUSIONS These findings establish α5β1 integrin as a pivotal driver of PAH pathology and we propose its inhibition as a novel, safe, and effective therapeutic strategy for PAH.
Collapse
Affiliation(s)
- Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Mónica S. Montesinos
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Yann Grobs
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Tetsuro Yokokawa
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Cardiovascular Medicine, Fukushima Medical University, Japan (T.Y.)
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Anesthesiology, St Mary’s Hospital, Kurume, Fukuoka, Japan (T.S.)
| | - Manon Mougin
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Charlotte Romanet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Mélanie Sauvaget
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Charlie Théberge
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Andréanne Pelletier
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Reem El Kabbout
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Keiko Yamamoto
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Muzaffar Akram
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Adrian S. Ray
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Blaise Lippa
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Bryan Goodwin
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Hua Wang
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - James E. Dowling
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Min Lu
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Qi Qiao
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - T. Andrew McTeague
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Terence I. Moy
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - François Potus
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| |
Collapse
|
4
|
Reuning U, D'Amore VM, Hodivala-Dilke K, Marinelli L, Kessler H. Importance of integrin transmembrane helical interactions for antagonistic versus agonistic ligand behavior: Consequences for medical applications. Bioorg Chem 2025; 156:108193. [PMID: 39842299 DOI: 10.1016/j.bioorg.2025.108193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Integrins are well-characterized receptors involved in cell adhesion and signaling. With six approved drugs, they are recognized as valuable therapeutic targets. Here, we explore potential activation mechanisms that may clarify the agonist versus antagonist behavior of integrin ligands. The reorganization of the transmembrane domain (TMD) in the integrin receptor, forming homooligomers within focal adhesions, could be key to the understanding of the agonistic properties of integrin ligands at substoichiometric concentrations. This has significant implications for medical applications. While we focus on the RGD peptide-recognizing integrin subfamily, we propose that these mechanistic insights may also apply to other integrin subtypes. For application of integrin ligands in medicine it is essential to consider this mechanism and its consequences for affinity and bioavailability.
Collapse
Affiliation(s)
- Ute Reuning
- TUM University Hospital, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Department of Gynecology and Obstetrics, Clinical Research Unit, Ismaninger Strasse 22, 81675 Munich, Germany.
| | - Vincenzo Maria D'Amore
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | - Luciana Marinelli
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Horst Kessler
- Institute for Advanced Study, Department of Chemistry, School of Natural Sciences and Bavarian NMR Center (BNMRZ), Technical University Munich, Ernst-Otto-Fischer-Str. 2, 85748 Garching, Germany.
| |
Collapse
|
5
|
Harrison BA, Dowling JE, Bursavich MG, Troast DM, Chong KM, Hahn KN, Zhong C, Mulvihill KM, Nguyen H, Monroy MF, Qiao Q, Sosa B, Mostafavi S, Smukste I, Lee D, Cappellucci L, Konopka EH, Nowakowski P, Stawski L, Senices M, Nguyen MH, Kapoor PS, Luus L, Sullivan A, Bortolato A, Svensson M, Hickey ER, Konze KD, Day T, Kim B, Negri A, Gerasyuto AI, Moy TI, Lu M, Ray AS, Wang L, Cui D, Lin FY, Lippa B, Rogers BN. The Discovery of MORF-627, a Highly Selective Conformationally-Biased Zwitterionic Integrin αvβ6 Inhibitor for Fibrosis. J Med Chem 2024; 67:18656-18681. [PMID: 39446989 DOI: 10.1021/acs.jmedchem.4c01851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Inhibition of integrin αvβ6 is a promising approach to the treatment of fibrotic disease such as idiopathic pulmonary fibrosis. Screening a small library combining head groups that stabilize the bent-closed conformation of integrin αIIbβ3 with αv integrin binding motifs resulted in the identification of hit compounds that bind the bent-closed conformation of αvβ6. Crystal structures of these compounds bound to αvβ6 and related integrins revealed opportunities to increase potency and selectivity, and these efforts were accelerated using accurate free energy perturbation (FEP+) calculations. Optimization of PK parameters including permeability, bioavailability, clearance, and half-life resulted in the discovery of development candidate MORF-627, a highly selective inhibitor of αvβ6 that stabilizes the bent-closed conformation and has good oral PK. Unfortunately, the compound showed toxicity in a 28-day NHP safety study, precluding further development. Nevertheless, MORF-627 is a useful tool compound for studying the biology of integrin αvβ6.
Collapse
Affiliation(s)
- Bryce A Harrison
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - James E Dowling
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | | | - Dawn M Troast
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Katherine M Chong
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Kristopher N Hahn
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Cheng Zhong
- Chemistry, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | | | - Hanh Nguyen
- CMC, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Meghan F Monroy
- Molecular and Cellular Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Qi Qiao
- Molecular and Cellular Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Brian Sosa
- Molecular and Cellular Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Siavash Mostafavi
- Molecular and Cellular Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Inese Smukste
- DMPK, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Dooyoung Lee
- DMPK, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Laura Cappellucci
- Screening Biology, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Elizabeth H Konopka
- Screening Biology, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Patrycja Nowakowski
- Screening Biology, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Lukasz Stawski
- Fibrosis, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Mayra Senices
- Fibrosis, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Minh Hai Nguyen
- Fibrosis, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Parmita S Kapoor
- Fibrosis, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Lia Luus
- Translational Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Andrew Sullivan
- Translational Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Andrea Bortolato
- Computational Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Mats Svensson
- Computational Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Eugene R Hickey
- Medicinal Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Kyle D Konze
- Medicinal Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Tyler Day
- Computational Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Byungchan Kim
- Computational Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Ana Negri
- Computational Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Aleksey I Gerasyuto
- Medicinal Chemistry, Therapeutics Group, Schrödinger, New York, New York 10036, United States
| | - Terence I Moy
- Screening Biology, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Min Lu
- Fibrosis, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Adrian S Ray
- Biology and Translational Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Liangsu Wang
- Biology and Translational Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Dan Cui
- DMPK, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Fu-Yang Lin
- Molecular and Cellular Sciences, Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Blaise Lippa
- Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| | - Bruce N Rogers
- Morphic Therapeutic, Waltham, Massachusetts 02451, United States
| |
Collapse
|
6
|
Jin M, Seed RI, Cai G, Shing T, Wang L, Ito S, Cormier A, Wankowicz SA, Jespersen JM, Baron JL, Carey ND, Campbell MG, Yu Z, Tang PK, Cossio P, Wen W, Lou J, Marks J, Nishimura SL, Cheng Y. Dynamic allostery drives autocrine and paracrine TGF-β signaling. Cell 2024; 187:6200-6219.e23. [PMID: 39288764 PMCID: PMC11531391 DOI: 10.1016/j.cell.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
TGF-β, essential for development and immunity, is expressed as a latent complex (L-TGF-β) non-covalently associated with its prodomain and presented on immune cell surfaces by covalent association with GARP. Binding to integrin αvβ8 activates L-TGF-β1/GARP. The dogma is that mature TGF-β must physically dissociate from L-TGF-β1 for signaling to occur. Our previous studies discovered that αvβ8-mediated TGF-β autocrine signaling can occur without TGF-β1 release from its latent form. Here, we show that mice engineered to express TGF-β1 that cannot release from L-TGF-β1 survive without early lethal tissue inflammation, unlike those with TGF-β1 deficiency. Combining cryogenic electron microscopy with cell-based assays, we reveal a dynamic allosteric mechanism of autocrine TGF-β1 signaling without release where αvβ8 binding redistributes the intrinsic flexibility of L-TGF-β1 to expose TGF-β1 to its receptors. Dynamic allostery explains the TGF-β3 latency/activation mechanism and why TGF-β3 functions distinctly from TGF-β1, suggesting that it broadly applies to other flexible cell surface receptor/ligand systems.
Collapse
Affiliation(s)
- Mingliang Jin
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Robert I Seed
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Guoqing Cai
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Tiffany Shing
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Li Wang
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Saburo Ito
- Department of Pathology, UCSF, San Francisco, CA, USA
| | | | | | | | - Jody L Baron
- Department of Medicine and UCSF Liver Center, UCSF, San Francisco, CA, USA
| | - Nicholas D Carey
- Department of Medicine and UCSF Liver Center, UCSF, San Francisco, CA, USA
| | - Melody G Campbell
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Zanlin Yu
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Phu K Tang
- Center for Computational Mathematics, Flatiron Institute, New York, NY, USA
| | - Pilar Cossio
- Center for Computational Mathematics, Flatiron Institute, New York, NY, USA; Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Weihua Wen
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | - James Marks
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | | | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA; Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA.
| |
Collapse
|
7
|
Marafi F, Esmail AA, Alfeeli MA, Sadeq A. 68 Ga-Trivehexin PET/CT in Metastatic Non-Small Cell Lung Cancer to the Brain. Clin Nucl Med 2024; 49:971-972. [PMID: 39190399 DOI: 10.1097/rlu.0000000000005406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
ABSTRACT In the era of molecular imaging and eager to study tumor tissues' microenvironment with noninvasive means, the search and development of new radiotracer targeted molecule continue. αvβ6-Integrin is a heterodimeric glycoprotein transmembrane receptor that is unique in that it is expressed exclusively in epithelial cells. It is upregulated in varieties of carcinomas such of the lung, breast, and colon. It plays a role in facilitating invasion, inhibiting apoptosis, regulating expression of matrix metalloproteases, and activating TGF-β in carcinoma. Expression of αvβ6 indicates poor prognosis and can help in development of targeted therapy. 68 Ga-Trivehexin has affinity of 85%-88% of this integrin.
Collapse
Affiliation(s)
- Fahad Marafi
- From the Department of Nuclear Medicine, Jaber Al Ahmed Center of Molecular Imaging
| | | | - Mahmoud A Alfeeli
- From the Department of Nuclear Medicine, Jaber Al Ahmed Center of Molecular Imaging
| | - Alyaa Sadeq
- From the Department of Nuclear Medicine, Jaber Al Ahmed Center of Molecular Imaging
| |
Collapse
|
8
|
Li F, Wu S, Lv L, Huang S, Zhang Z, Zerang Z, Li P, Cao Y, Bao H, Sun P, Bai X, He Y, Fu Y, Yuan H, Ma X, Zhao Z, Zhang J, Wang J, Wang T, Li D, Zhang Q, He J, Liu Z, Lu Z, Lei D, Li K. Discovery, recognized antigenic structures, and evolution of cross-serotype broadly neutralizing antibodies from porcine B-cell repertoires against foot-and-mouth disease virus. PLoS Pathog 2024; 20:e1012623. [PMID: 39405339 PMCID: PMC11508087 DOI: 10.1371/journal.ppat.1012623] [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: 03/04/2024] [Revised: 10/25/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
It is a great challenge to isolate the broadly neutralizing antibodies (bnAbs) against foot-and-mouth disease virus (FMDV) due to its existence as seven distinct serotypes without cross-protection. Here, by vaccination of pig with FMDV serotypes O and A whole virus antigens, we obtained 10 bnAbs against serotypes O, A and/or Asia1 by dissecting 216 common clonotypes of two serotypes O and A specific porcine B-cell receptor (BCR) gene repertoires containing total 12720 B cell clones, indicating the induction of cross-serotype bnAbs after sequential vaccination with serotypes O and A antigens. The majority of porcine bnAbs (9/10) were derived from terminally differentiated B cells of different clonal lineages, which convergently targeted the conserved "RGDL" motif on structural protein VP1 of FMDV by mimicking receptor recognition to inhibit viral attachment to cells. Cryo-EM complex structures revealed that the other bnAb pOA-2 specifically targets a novel inter-pentamer antigen structure surrounding the viral three-fold axis, with a highly conserved determinant at residue 68 on VP2. This unique binding pattern enabled cross-serotype neutralization by destabilizing the viral particle. The evolutionary analysis of pOA-2 demonstrated its origin from an intermediate B-cell, emphasizing the crucial role of somatic hypermutations (SHMs) in balancing the breadth and potency of neutralization. However, excessive SHMs may deviate from the trajectory of broad neutralization. This study provides a strategy to uncover bnAbs against highly mutable pathogens and the cross-serotype antigenic structures to explore broadly protective FMDV vaccine.
Collapse
Affiliation(s)
- Fengjuan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Shanquan Wu
- School of Physical Science and Technology, Electron Microscopy Centre of Lanzhou University, Lanzhou University, Lanzhou, P. R. China
| | - Lv Lv
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Shulun Huang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Zelin Zhang
- School of Physical Science and Technology, Electron Microscopy Centre of Lanzhou University, Lanzhou University, Lanzhou, P. R. China
| | - Zhaxi Zerang
- School of Physical Science and Technology, Electron Microscopy Centre of Lanzhou University, Lanzhou University, Lanzhou, P. R. China
| | - Pinghua Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Yimei Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Huifang Bao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Pu Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Xingwen Bai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Yong He
- School of Pharmaceutical Sciences, Shandong University, Ji’nan, P. R. China
| | - Yuanfang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Hong Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Xueqing Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Zhixun Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Jian Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Dong Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Qiang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Jijun He
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Zaixin Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Zengjun Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| | - Dongsheng Lei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- School of Physical Science and Technology, Electron Microscopy Centre of Lanzhou University, Lanzhou University, Lanzhou, P. R. China
| | - Kun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P. R. China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, P. R. China
| |
Collapse
|
9
|
Vu Hong A, Suel L, Petat E, Dubois A, Le Brun PR, Guerchet N, Veron P, Poupiot J, Richard I. An engineered AAV targeting integrin alpha V beta 6 presents improved myotropism across species. Nat Commun 2024; 15:7965. [PMID: 39261465 PMCID: PMC11390886 DOI: 10.1038/s41467-024-52002-4] [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: 10/25/2023] [Accepted: 08/22/2024] [Indexed: 09/13/2024] Open
Abstract
Current adeno-associated virus (AAV) gene therapy using nature-derived AAVs is limited by non-optimal tissue targeting. In the treatment of muscular diseases (MD), high doses are often required but can lead to severe adverse effects. Here, we rationally design an AAV capsid that specifically targets skeletal muscle to lower treatment doses. We computationally integrate binding motifs of human integrin alphaV beta6, a skeletal muscle receptor, into a liver-detargeting capsid. Designed AAVs show higher productivity and superior muscle transduction compared to their parent. One variant, LICA1, demonstrates comparable muscle transduction to other myotropic AAVs with reduced liver targeting. LICA1's myotropic properties are observed across species, including non-human primate. Consequently, LICA1, but not AAV9, effectively delivers therapeutic transgenes and improved muscle functionality in two mouse MD models (male mice) at a low dose (5E12 vg/kg). These results underline the potential of our design method for AAV engineering and LICA1 variant for MD gene therapy.
Collapse
Affiliation(s)
- Ai Vu Hong
- Genethon, 1 bis rue de l'internationale, Evry, France.
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France.
| | - Laurence Suel
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Eva Petat
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Auriane Dubois
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Pierre-Romain Le Brun
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Nicolas Guerchet
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Philippe Veron
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Jérôme Poupiot
- Genethon, 1 bis rue de l'internationale, Evry, France
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France
| | - Isabelle Richard
- Genethon, 1 bis rue de l'internationale, Evry, France.
- INTEGRARE research unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), Evry, France.
- Atamyo Therapeutics, 1 bis rue de l'internationale, Evry, France.
| |
Collapse
|
10
|
Bellani S, Molyneaux PL, Maher TM, Spagnolo P. Potential of αvβ6 and αvβ1 integrin inhibition for treatment of idiopathic pulmonary fibrosis. Expert Opin Ther Targets 2024; 28:575-585. [PMID: 38949181 DOI: 10.1080/14728222.2024.2375375] [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: 03/23/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease of unknown cause with a dismal prognosis. Nintedanib and Pirfenidone are approved worldwide for the treatment of IPF, but they only slow the rate of functional decline and disease progression. Therefore, there is an urgent need for more efficacious and better tolerated drugs. AREAS COVERED αvβ6 and αvβ1 are two integrins overexpressed in fibrotic tissue, which play a critical role in the development of lung fibrosis. They act by converting transforming growth factor (TGF)-β, one of the most important profibrotic cytokine, in its active form. Here, we summarize and critically discuss the potential of a dual αvβ6/αvβ1 integrin inhibitor for the treatment of IPF. EXPERT OPINION Bexotegrast, a dual αvβ6/αvβ1 integrin inhibitor, has the potential to slow or even halt disease progression in IPF. Indeed, the strong pre-clinical rationale and promising early phase clinical trial data have raised expectations.
Collapse
Affiliation(s)
- Serena Bellani
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College, London, UK
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Hospitals, London, UK
| | - Toby M Maher
- Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
11
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
12
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
13
|
Zhang Z, Wang Z, Liu T, Tang J, Liu Y, Gou T, Chen K, Wang L, Zhang J, Yang Y, Zhang H. Exploring the role of ITGB6: fibrosis, cancer, and other diseases. Apoptosis 2024; 29:570-585. [PMID: 38127283 DOI: 10.1007/s10495-023-01921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Integrin β6 (ITGB6), a member of the integrin family of proteins, is only present in epithelial tissues and frequently associates with integrin subunit αv to form transmembrane heterodimers named integrin αvβ6. Importantly, ITGB6 determines αvβ6 expression and availability. In addition to being engaged in organ fibrosis, ITGB6 is also directly linked to the emergence of cancer, periodontitis, and several potential genetic diseases. Therefore, it is of great significance to study the molecular-biological mechanism of ITGB6, which could provide novel insights for future clinical diagnosis and therapy. This review introduces the structure, distribution, and biological function of ITGB6. This review also expounds on ITGB6-related diseases, detailing the known biological effects of ITGB6.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Tong Liu
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yanqing Liu
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Tiantian Gou
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Kangli Chen
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Li Wang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Juan Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Huan Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
14
|
Lemay SE, Montesinos MS, Grobs Y, Yokokawa T, Shimauchi T, Romanet C, Sauvaget M, Breuils-Bonnet S, Bourgeois A, Théberge C, Pelletier A, El Kabbout R, Martineau S, Yamamoto K, Ray AS, Lippa B, Goodwin B, Lin FY, Wang H, Dowling JE, Lu M, Qiao Q, McTeague TA, Moy TI, Potus F, Provencher S, Boucherat O, Bonnet S. Exploring Integrin α5β1 as a Potential Therapeutic Target for Pulmonary Arterial Hypertension: Insights from Comprehensive Multicenter Preclinical Studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596052. [PMID: 38854025 PMCID: PMC11160677 DOI: 10.1101/2024.05.27.596052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by obliterative vascular remodeling of the small pulmonary arteries (PA) and progressive increase in pulmonary vascular resistance (PVR) leading to right ventricular (RV) failure. Although several drugs are approved for the treatment of PAH, mortality remains high. Accumulating evidence supports a pathological function of integrins in vessel remodeling, which are gaining renewed interest as drug targets. However, their role in PAH remains largely unexplored. We found that the arginine-glycine-aspartate (RGD)-binding integrin α5β1 is upregulated in PA endothelial cells (PAEC) and PA smooth muscle cells (PASMC) from PAH patients and remodeled PAs from animal models. Blockade of the integrin α5β1 or depletion of the α5 subunit resulted in mitotic defects and inhibition of the pro-proliferative and apoptosis-resistant phenotype of PAH cells. Using a novel small molecule integrin inhibitor and neutralizing antibodies, we demonstrated that α5β1 integrin blockade attenuates pulmonary vascular remodeling and improves hemodynamics and RV function in multiple preclinical models. Our results provide converging evidence to consider α5β1 integrin inhibition as a promising therapy for pulmonary hypertension. One sentence summary The α5β1 integrin plays a crucial role in pulmonary vascular remodeling.
Collapse
|
15
|
Yamada A, Takei T, Kawakami T, Taniguchi Y, Sekiguchi K, Hojo H. Application of cysteinyl prolyl ester for the synthesis of cyclic peptides containing an RGD sequence and their biological activity measurement. Front Chem 2024; 12:1391678. [PMID: 38873405 PMCID: PMC11169864 DOI: 10.3389/fchem.2024.1391678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Cysteinyl RGD-peptidyl cysteinyl prolyl esters, which have different configurations at the cysteine and proline residues, were synthesized by the solid-phase method and cyclized by the native chemical ligation reaction. Cyclization efficiently proceeded to give cyclic peptides, regardless of the difference in the configuration. The peptides were further derivatized to the corresponding desulfurized or methylated cyclic peptides at the Cys residues. The inhibition activity to αvβ6 integrin binding was then analyzed by ELISA. The results showed that the activity varied depending on the difference in the configuration and modification of the cysteinyl prolyl ester (CPC) moiety, demonstrating the usefulness of this method in the search for a good inhibitor of the protein-protein interaction.
Collapse
Affiliation(s)
| | | | | | | | | | - Hironobu Hojo
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
16
|
Liao J, Chen R, Lin B, Deng R, Liang Y, Zeng J, Ma S, Qiu X. Cross-Talk between the TGF-β and Cell Adhesion Signaling Pathways in Cancer. Int J Med Sci 2024; 21:1307-1320. [PMID: 38818471 PMCID: PMC11134594 DOI: 10.7150/ijms.96274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
Transforming growth factor-β (TGF-β) is strongly associated with the cell adhesion signaling pathway in cell differentiation, migration, etc. Mechanistically, TGF-β is secreted in an inactive form and localizes to the extracellular matrix (ECM) via the latent TGF-β binding protein (LTBP). However, it is the release of mature TGF-β that is essential for the activation of the TGF-β signaling pathway. This progress requires specific integrins (one of the main groups of cell adhesion molecules (CAMs)) to recognize and activate the dormant TGF-β. In addition, TGF-β regulates cell adhesion ability through modulating CAMs expression. The aberrant activation of the TGF-β signaling pathway, caused by abnormal expression of key regulatory molecules (such as Smad proteins, certain transcription factors, and non-coding RNAs), promotes tumor invasive and metastasis ability via epithelial-mesenchymal transition (EMT) during the late stages of tumorigenesis. In this paper, we summarize the crosstalk between TGF-β and cell adhesion signaling pathway in cancer and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Jiahao Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Rentang Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Runhua Deng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Yanfang Liang
- Department of Pathology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong, 523905, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Sha Ma
- School of Biomedical Engineering, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| |
Collapse
|
17
|
Brown NF, Murray ER, Cutmore LC, Howard P, Masterson L, Zammarchi F, Hartley JA, van Berkel PH, Marshall JF. Integrin-αvβ6 targeted peptide-toxin therapy in a novel αvβ6-expressing immunocompetent model of pancreatic cancer. Pancreatology 2024; 24:445-455. [PMID: 38519394 DOI: 10.1016/j.pan.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/24/2024]
Abstract
Previously we reported that a novel αvβ6-specific peptide-drug conjugate (SG3299) could eliminate established human pancreatic ductal adenocarcinoma (PDAC) xenografts. However the development of effective therapies for PDAC, which is an essential need, must show efficacy in relevant immunocompetent animals. Previously we reported that the KPC mouse transgenic PDAC model that closely recapitulates most stages of development of human PDAC, unlike in humans, failed to express αvβ6 on their tumours or metastases. In this study we have taken the KPC-derived PDAC line TB32043 and engineered a variant line (TB32043mb6S2) that expresses mouse integrin αvβ6. We report that orthotopic implantation of the αvβ6 over-expressing TB32043mb6S2 cells promotes shorter overall survival and increase in metastases. Moreover, systemic treatment of mice with established TB32043mb6S2 tumours in the pancreas with SG2399 lived significantly longer (p < 0.001; mean OS 48d) compared with PBS or control SG3511 (mean OS 25.5d and 26d, respectively). Thus SG3299 is confirmed as a promising candidate therapeutic for the therapy of PDAC.
Collapse
Affiliation(s)
- Nicholas F Brown
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Elizabeth R Murray
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Lauren C Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Philip Howard
- Spirogen, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Luke Masterson
- Spirogen, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Francesca Zammarchi
- ADC Therapeutics (UK) Ltd, Translation & Innovation Hub Building, Imperial College White City Campus, 84 Wood Lane, London, W12 0BZ, UK
| | - John A Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - Patrick H van Berkel
- ADC Therapeutics (UK) Ltd, Translation & Innovation Hub Building, Imperial College White City Campus, 84 Wood Lane, London, W12 0BZ, UK
| | - John F Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
18
|
Cao Y, Su H, Zeng J, Xie Y, Liu Z, Liu F, Qiu Y, Yi F, Lin J, Hammes HP, Zhang C. Integrin β8 prevents pericyte-myofibroblast transition and renal fibrosis through inhibiting the TGF-β1/TGFBR1/Smad3 pathway in diabetic kidney disease. Transl Res 2024; 265:36-50. [PMID: 37931653 DOI: 10.1016/j.trsl.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023]
Abstract
Diabetic kidney disease (DKD) is one of the leading causes to develop end-stage kidney disease worldwide. Pericytes are implicated in the development of tissue fibrosis. However, the underlying mechanisms of pericytes in DKD remain largely unknown. We isolated and cultured primary pericytes and rat mesangial cells (HBZY-1). Western blot and qRT-PCR analysis were used to explore the role and regulatory mechanism of Integrin β8/transforming growth factor beta 1 (TGF-β1) pathway. We also constructed pericyte-specific Integrin β8 knock-in mice as the research objects to determine the role of Integrin β8 in vivo. We discovered that reduced Integrin β8 expression was closely associated with pericyte transition in DKD. Overexpressed Integrin β8 in pericytes dramatically suppressed TGF-β1/TGF beta receptor 1 (TGFBR1)/Smad3 signaling pathway and protected glomerular endothelial cells (GECs) in vitro. In vivo, pericyte-specific Integrin β8 knock-in ameliorated pericyte transition, endothelium injury and renal fibrosis in STZ-induced diabetic mice. Mechanistically, Murine double minute 2 (MDM2) was found to increase the degradation of Integrin β8 and caused TGF-β1 release and activation. Knockdown MDM2 could partly reverse the decline of Integrin β8 and suppress pericytes transition. In conclusion, the present findings suggested that upregulated MDM2 expression contributes to the degradation of Integrin β8 and activation of TGF-β1/TGFBR1/Smad3 signaling pathway, which ultimately leads to pericyte transition during DKD progression. These results indicate MDM2/Integrin β8 might be considered as therapeutic targets for DKD.
Collapse
Affiliation(s)
- Yiling Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jieyu Zeng
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaru Xie
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zezhou Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Qiu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jihong Lin
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
19
|
Hao Y, Yan J, Fraser C, Jiang A, Anuganti M, Zhang R, Lloyd K, Jardine J, Coppola J, Meijers R, Li J, Springer TA. Synthetic integrin antibodies discovered by yeast display reveal αV subunit pairing preferences with β subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577394. [PMID: 38328192 PMCID: PMC10849667 DOI: 10.1101/2024.01.26.577394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Eight of the 24 integrin heterodimers bind to the tripeptide Arg-Gly-Asp (RGD) motif in their extracellular ligands, and play essential roles in cell adhesion, migration, and homeostasis. Despite similarity in recognizing the RGD motif and some redundancy, these integrins can selectively recognize RGD-containing ligands including fibronectin, vitronectin, fibrinogen, nephronectin and the prodomain of the transforming growth factors to fulfill specific functions in cellular processes. Subtype-specific antibodies against RGD-binding integrins are desirable for investigating their specific functions. In this study, we discovered 11 antibodies that exhibit high specificity and affinity towards integrins αVβ3, αVβ5, αVβ6, αVβ8, and α5β1 from a synthetic yeast-displayed Fab library. Of these, 6 are function-blocking antibodies containing an R(G/L/T) D motif in their CDR3 sequences. We report antibody binding specificity, kinetics, and binding affinity for purified integrin ectodomains as well as intact integrins on the cell surface. We further employed these antibodies to reveal binding preferences of the αV subunit for its 5 β-subunit partners: β6=β8>β3>β1=β5.
Collapse
Affiliation(s)
- Yuxin Hao
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Courtney Fraser
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Murali Anuganti
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Roushu Zhang
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Kenneth Lloyd
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Joseph Jardine
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Jessica Coppola
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Rob Meijers
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Jing Li
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Hao Y, Yan J, Fraser C, Jiang A, Anuganti M, Zhang R, Lloyd K, Jardine J, Coppola J, Meijers R, Li J, Springer TA. Synthetic integrin antibodies discovered by yeast display reveal αV subunit pairing preferences with β subunits. MAbs 2024; 16:2365891. [PMID: 38889315 PMCID: PMC11188837 DOI: 10.1080/19420862.2024.2365891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Integrins are cell surface receptors that mediate the interactions of cells with their surroundings and play essential roles in cell adhesion, migration, and homeostasis. Eight of the 24 integrins bind to the tripeptide Arg-Gly-Asp (RGD) motif in their extracellular ligands, comprising the RGD-binding integrin subfamily. Despite similarity in recognizing the RGD motif and some redundancy, these integrins can selectively recognize RGD-containing ligands to fulfill specific functions in cellular processes. Antibodies against individual RGD-binding integrins are desirable for investigating their specific functions, and were selected here from a synthetic yeast-displayed Fab library. We discovered 11 antibodies that exhibit high specificity and affinity toward their target integrins, i.e. αVβ3, αVβ5, αVβ6, αVβ8, and α5β1. Of these, six are function-blocking antibodies and contain a ligand-mimetic R(G/L/T)D motif in their CDR3 sequences. We report antibody-binding specificity, kinetics, and binding affinity for purified integrin ectodomains, as well as intact integrins on the cell surface. We further used these antibodies to reveal binding preferences of the αV subunit for its 5 β-subunit partners: β6 = β8 > β3 > β1 = β5.
Collapse
Affiliation(s)
- Yuxin Hao
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Courtney Fraser
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Roushu Zhang
- Institute for Protein Innovation, Boston, MA, USA
| | | | | | | | - Rob Meijers
- Institute for Protein Innovation, Boston, MA, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Woodward EA, Wang E, Wallis C, Sharma R, Tie AWJ, Murthy N, Blancafort P. Protocol for Delivery of CRISPR/dCas9 Systems for Epigenetic Editing into Solid Tumors Using Lipid Nanoparticles Encapsulating RNA. Methods Mol Biol 2024; 2842:267-287. [PMID: 39012601 DOI: 10.1007/978-1-0716-4051-7_14] [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] [Indexed: 07/17/2024]
Abstract
Genome editing tools, particularly the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) systems (e.g., CRISPR/Cas9), and their repurposing into epigenetic editing platforms, offer enormous potential as safe and customizable therapies for cancer. Specifically, various transcriptional abnormalities in human malignancies, such as silencing of tumor suppressors and ectopic re-expression of oncogenes, have been successfully targeted with virtually no off-target effects using CRISPR activation and repression systems. In these systems, the nuclease-deactivated Cas9 protein (dCas9) is fused to one or more domains inducing selective activation or repression of the targeted genes. Despite these advances, the efficient in vivo delivery of these molecules into the target cancer cells represents a critical barrier to accomplishing translation into a clinical therapy setting for cancer. Major obstacles include the large size of dCas9 fusion proteins, the necessity of multimodal delivery of protein and gRNAs, and the potential of these formulations to elicit detrimental immune responses.In this context, viral methods for delivering CRISPR face several limitations, such as the packaging capacity of the viral genome, the potential for integration of the nucleic acids into the host cells genome, and immunogenicity of viral proteins, posing serious safety concerns. The rapid development of mRNA vaccines in response to the COVID-19 pandemic has rekindled interest in mRNA-based approaches for CRISPR/dCas9 delivery. Simultaneously, due to their high loading capacity, scalability, customizable surface modification for cell targeting, and low immunogenicity, lipid nanoparticles (LNPs) have been widely explored as nonviral vectors. In this chapter, we first describe the design of optimized dCas9-effector mRNAs and gRNAs for epigenetic editing. We outline formulations of LNPs suitable for dCas9 mRNA delivery. Additionally, we provide a protocol for the co-encapsulation of the dCas9-effector mRNAs and gRNA into these LNPs, along with detailed methods for delivering these formulations to both cell lines (in vitro) and mouse models of breast cancer (in vivo).
Collapse
Affiliation(s)
- Eleanor A Woodward
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Edina Wang
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Christopher Wallis
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Rohit Sharma
- Department of Bioengineering, University of California, Berkeley, CA, USA
- The Innovative Genomics Institute, Berkeley, CA, USA
| | - Ash W J Tie
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, CA, USA
- The Innovative Genomics Institute, Berkeley, CA, USA
| | - Pilar Blancafort
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA, Australia.
- Centre for Medical Research, University of Western Australia, Perth, WA, Australia.
- The Innovative Genomics Institute, Berkeley, CA, USA.
| |
Collapse
|
22
|
D’Amore V, Donati G, Lenci E, Ludwig BS, Kossatz S, Baiula M, Trabocchi A, Kessler H, Di Leva FS, Marinelli L. Molecular View on the iRGD Peptide Binding Mechanism: Implications for Integrin Activity and Selectivity Profiles. J Chem Inf Model 2023; 63:6302-6315. [PMID: 37788340 PMCID: PMC10598797 DOI: 10.1021/acs.jcim.3c01071] [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: 07/14/2023] [Indexed: 10/05/2023]
Abstract
Receptor-selective peptides are widely used as smart carriers for specific tumor-targeted delivery. A remarkable example is the cyclic nonapeptide iRGD (CRGDKPGDC, 1) that couples intrinsic cytotoxic effects with striking tumor-homing properties. These peculiar features are based on a rather complex multistep mechanism of action, where the primary event is the recognition of RGD integrins. Despite the high number of preclinical studies and the recent success of a phase I trial for the treatment of pancreatic ductal adenocarcinoma (PDAC), there is little information available about the iRGD three-dimensional (3D) structure and integrin binding properties. Here, we re-evaluate the peptide's affinity for cancer-related integrins including not only the previously known targets αvβ3 and αvβ5 but also the αvβ6 isoform, which is known to drive cell growth, migration, and invasion in many malignancies including PDAC. Furthermore, we use parallel tempering in the well-tempered ensemble (PT-WTE) metadynamics simulations to characterize the in-solution conformation of iRGD and extensive molecular dynamics calculations to fully investigate its binding mechanism to integrin partners. Finally, we provide clues for fine-tuning the peptide's potency and selectivity profile, which, in turn, may further improve its tumor-homing properties.
Collapse
Affiliation(s)
- Vincenzo
Maria D’Amore
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Greta Donati
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Elena Lenci
- Department
of Chemistry “Ugo Schiff″, University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Florence, Italy
| | - Beatrice Stefanie Ludwig
- Department
of Nuclear Medicine, University Hospital Klinikum Rechts der Isar
and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, Munich 81675, Germany
| | - Susanne Kossatz
- Department
of Nuclear Medicine, University Hospital Klinikum Rechts der Isar
and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, Munich 81675, Germany
- Department
of Chemistry, Institute for Advanced Study, Technical University Munich, Garching 85748, Germany
| | - Monica Baiula
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Andrea Trabocchi
- Department
of Chemistry “Ugo Schiff″, University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Florence, Italy
| | - Horst Kessler
- Department
of Chemistry, Institute for Advanced Study, Technical University Munich, Garching 85748, Germany
| | - Francesco Saverio Di Leva
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, Università degli Studi
di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
23
|
Zhang H, Zhu DS, Zhu J. Family-wide analysis of integrin structures predicted by AlphaFold2. Comput Struct Biotechnol J 2023; 21:4497-4507. [PMID: 37753178 PMCID: PMC10518446 DOI: 10.1016/j.csbj.2023.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/17/2023] [Accepted: 09/17/2023] [Indexed: 09/28/2023] Open
Abstract
Recent advances in protein structure prediction using AlphaFold2, known for its high efficiency and accuracy, have opened new avenues for comprehensive analysis of all structures within a single protein family. In this study, we evaluated the capabilities of AphaFold2 in analyzing integrin structures. Integrins are heterodimeric cell surface receptors composed of a combination of 18 α and 8 β subunits, resulting in a family of 24 different members. Both α and β subunits consist of a large extracellular domain, a short transmembrane domain, and typically, a short cytoplasmic tail. Integrins play a pivotal role in a wide range of cellular functions by recognizing diverse ligands. Despite significant advances in integrin structural studies in recent decades, high-resolution structures have only been determined for a limited subsets of integrin members, thus limiting our understanding of the entire integrin family. Here, we first analyzed the single-chain structures of 18 α and 8 β integrins in the AlphaFold2 protein structure database. We then employed the newly developed AlphaFold2-multimer program to predict the α/β heterodimer structures of all 24 human integrins. The predicted structures show a high level of accuracy for the subdomains of both α and β subunits, offering high-resolution structure insights for all integrin heterodimers. Our comprehensive structural analysis of the entire integrin family unveils a potentially diverse range of conformations among the 24 members, providing a valuable structure database for studies related to integrin structure and function. We further discussed the potential applications and limitations of the AlphaFold2-derived integrin structures.
Collapse
Affiliation(s)
- Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Daniel S. Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
24
|
Zhang H, Zhu DS, Zhu J. Family-wide analysis of integrin structures predicted by AlphaFold2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539023. [PMID: 37205578 PMCID: PMC10187181 DOI: 10.1101/2023.05.02.539023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Recent advances in protein structure prediction using AlphaFold2, known for its high efficiency and accuracy, have opened new avenues for comprehensive analysis of all structures within a single protein family. In this study, we evaluated the capabilities of AphaFold2 in analyzing integrin structures. Integrins are heterodimeric cell surface receptors composed of a combination of 18 α and 8 β subunits, resulting in a family of 24 different members. Both α and β subunits consist of a large extracellular domain, a short transmembrane domain, and typically, a short cytoplasmic tail. Integrins play a pivotal role in a wide range of cellular functions by recognizing diverse ligands. Despite significant advances in integrin structural studies in recent decades, high-resolution structures have only been determined for a limited subsets of integrin members, thus limiting our understanding of the entire integrin family. Here, we first analyzed the single-chain structures of 18 α and 8 β integrins in the AlphaFold2 protein structure database. We then employed the newly developed AlphaFold2-multimer program to predict the α/β heterodimer structures of all 24 human integrins. The predicted structures show a high level of accuracy for the subdomains of both α and β subunits, offering high-resolution structure insights for all integrin heterodimers. Our comprehensive structural analysis of the entire integrin family unveils a potentially diverse range of conformations among the 24 members, providing a valuable structure database for studies related to integrin structure and function. We further discussed the potential applications and limitations of the AlphaFold2-derived integrin structures.
Collapse
Affiliation(s)
- Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Daniel S. Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
Roy A, Shi L, Chang A, Dong X, Fernandez A, Kraft JC, Li J, Le VQ, Winegar RV, Cherf GM, Slocum D, Poulson PD, Casper GE, Vallecillo-Zúniga ML, Valdoz JC, Miranda MC, Bai H, Kipnis Y, Olshefsky A, Priya T, Carter L, Ravichandran R, Chow CM, Johnson MR, Cheng S, Smith M, Overed-Sayer C, Finch DK, Lowe D, Bera AK, Matute-Bello G, Birkland TP, DiMaio F, Raghu G, Cochran JR, Stewart LJ, Campbell MG, Van Ry PM, Springer T, Baker D. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. Nat Commun 2023; 14:5660. [PMID: 37704610 PMCID: PMC10500007 DOI: 10.1038/s41467-023-41272-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
The RGD (Arg-Gly-Asp)-binding integrins αvβ6 and αvβ8 are clinically validated cancer and fibrosis targets of considerable therapeutic importance. Compounds that can discriminate between homologous αvβ6 and αvβ8 and other RGD integrins, stabilize specific conformational states, and have high thermal stability could have considerable therapeutic utility. Existing small molecule and antibody inhibitors do not have all these properties, and hence new approaches are needed. Here we describe a generalized method for computationally designing RGD-containing miniproteins selective for a single RGD integrin heterodimer and conformational state. We design hyperstable, selective αvβ6 and αvβ8 inhibitors that bind with picomolar affinity. CryoEM structures of the designed inhibitor-integrin complexes are very close to the computational design models, and show that the inhibitors stabilize specific conformational states of the αvβ6 and the αvβ8 integrins. In a lung fibrosis mouse model, the αvβ6 inhibitor potently reduced fibrotic burden and improved overall lung mechanics, demonstrating the therapeutic potential of de novo designed integrin binding proteins with high selectivity.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lei Shi
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Encodia Inc, 5785 Oberlin Drive, San Diego, CA, 92121, USA
| | - Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Xianchi Dong
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China
| | - Andres Fernandez
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - John C Kraft
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Viet Q Le
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Rebecca Viazzo Winegar
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Gerald Maxwell Cherf
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Denali Therapeutics, South San Francisco, CA, USA
| | - Dean Slocum
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - P Daniel Poulson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Garrett E Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | | | - Jonard Corpuz Valdoz
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Marcos C Miranda
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hua Bai
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Yakov Kipnis
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Audrey Olshefsky
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tanu Priya
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Lauren Carter
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Cameron M Chow
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Max R Johnson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Suna Cheng
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - McKaela Smith
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Catherine Overed-Sayer
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K Finch
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Alchemab Therapeutics Ltd, Cambridge, UK
| | - David Lowe
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Evox Therapeutics Limited, Oxford Science Park, Medawar Centre, East Building, Robert Robinson Avenue, Oxford, OX4 4HG, England
| | - Asim K Bera
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, USA
| | - Timothy P Birkland
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Frank DiMaio
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Lance J Stewart
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| | - Pam M Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA.
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA.
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
26
|
Urquiza M, Benavides-Rubio D, Jimenez-Camacho S. Structural analysis of peptide binding to integrins for cancer detection and treatment. Biophys Rev 2023; 15:699-708. [PMID: 37681100 PMCID: PMC10480133 DOI: 10.1007/s12551-023-01084-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/20/2023] [Indexed: 09/09/2023] Open
Abstract
Integrins are cell receptors involved in several metabolic pathways often associated with cell proliferation. Some of these integrins are downregulated during human physical development, but when these integrins are overexpressed in adult humans, they can be associated with several diseases, such as cancer. Molecules that specifically bind to these integrins are useful for cancer detection, diagnosis, and treatment. This review focuses on the structures of integrin-peptidic ligand complexes to dissect how the binding occurs and the molecular basis of the specificity and affinity of these peptidic ligands. Understanding these interactions at the molecular level is fundamental to be able to design new peptides that are more specific and more sensitive to a particular integrin. The integrin complexes covered in this review are α5β1, αIIbβ3, αvβ3, αvβ6, and αvβ8, because the molecular structures of the complex have been experimentally determined and their presence on tumor cancer cells are associated with a poor prognosis, making them targets for cancer detection and treatment.
Collapse
Affiliation(s)
- Mauricio Urquiza
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| | - Daniela Benavides-Rubio
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| | - Silvia Jimenez-Camacho
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| |
Collapse
|
27
|
Le VQ, Zhao B, Ramesh S, Toohey C, DeCosta A, Mintseris J, Liu X, Gygi S, Springer TA. A specialized integrin-binding motif enables proTGF-β2 activation by integrin αVβ6 but not αVβ8. Proc Natl Acad Sci U S A 2023; 120:e2304874120. [PMID: 37279271 PMCID: PMC10268255 DOI: 10.1073/pnas.2304874120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023] Open
Abstract
Activation of latent transforming growth factor (TGF)-β2 is incompletely understood. Unlike TGF-β1 and β3, the TGF-β2 prodomain lacks a seven-residue RGDLXX (L/I) integrin-recognition motif and is thought not to be activated by integrins. Here, we report the surprising finding that TGF-β2 contains a related but divergent 13-residue integrin-recognition motif (YTSGDQKTIKSTR) that specializes it for activation by integrin αVβ6 but not αVβ8. Both classes of motifs compete for the same binding site in αVβ6. Multiple changes in the longer motif underlie its specificity. ProTGF-β2 structures define interesting differences from proTGF-β1 and the structural context for activation by αVβ6. Some integrin-independent activation is also seen for proTGF-β2 and even more so for proTGF-β3. Our findings have important implications for therapeutics to αVβ6 in clinical trials for fibrosis, in which inhibition of TGF-β2 activation has not been anticipated.
Collapse
Affiliation(s)
- Viet Q. Le
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| | - Bo Zhao
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| | - Siddanth Ramesh
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
| | - Cameron Toohey
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
| | - Adam DeCosta
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
| | - Julian Mintseris
- Department of Cell Biology, Harvard Medical School,Boston, MA02115
| | - Xinyue Liu
- Department of Cell Biology, Harvard Medical School,Boston, MA02115
| | - Steven Gygi
- Department of Cell Biology, Harvard Medical School,Boston, MA02115
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
28
|
Roy A, Shi L, Chang A, Dong X, Fernandez A, Kraft JC, Li J, Le VQ, Winegar RV, Cherf GM, Slocum D, Daniel Poulson P, Casper GE, Vallecillo-Zúniga ML, Valdoz JC, Miranda MC, Bai H, Kipnis Y, Olshefsky A, Priya T, Carter L, Ravichandran R, Chow CM, Johnson MR, Cheng S, Smith M, Overed-Sayer C, Finch DK, Lowe D, Bera AK, Matute-Bello G, Birkland TP, DiMaio F, Raghu G, Cochran JR, Stewart LJ, Campbell MG, Van Ry PM, Springer T, Baker D. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544624. [PMID: 37398153 PMCID: PMC10312613 DOI: 10.1101/2023.06.12.544624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The RGD (Arg-Gly-Asp)-binding integrins αvβ6 and αvβ8 are clinically validated cancer and fibrosis targets of considerable therapeutic importance. Compounds that can discriminate between the two closely related integrin proteins and other RGD integrins, stabilize specific conformational states, and have sufficient stability enabling tissue restricted administration could have considerable therapeutic utility. Existing small molecules and antibody inhibitors do not have all of these properties, and hence there is a need for new approaches. Here we describe a method for computationally designing hyperstable RGD-containing miniproteins that are highly selective for a single RGD integrin heterodimer and conformational state, and use this strategy to design inhibitors of αvβ6 and αvβ8 with high selectivity. The αvβ6 and αvβ8 inhibitors have picomolar affinities for their targets, and >1000-fold selectivity over other RGD integrins. CryoEM structures are within 0.6-0.7Å root-mean-square deviation (RMSD) to the computational design models; the designed αvβ6 inhibitor and native ligand stabilize the open conformation in contrast to the therapeutic anti-αvβ6 antibody BG00011 that stabilizes the bent-closed conformation and caused on-target toxicity in patients with lung fibrosis, and the αvβ8 inhibitor maintains the constitutively fixed extended-closed αvβ8 conformation. In a mouse model of bleomycin-induced lung fibrosis, the αvβ6 inhibitor potently reduced fibrotic burden and improved overall lung mechanics when delivered via oropharyngeal administration mimicking inhalation, demonstrating the therapeutic potential of de novo designed integrin binding proteins with high selectivity.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Lei Shi
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Current Address: Encodia Inc, 5785 Oberlin Drive, San Diego, CA 92121
| | - Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Xianchi Dong
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
- Current address: State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Engineering Research Center of Protein and Peptide Medicine,Ministry of Education
| | - Andres Fernandez
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - John C. Kraft
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Viet Q. Le
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Rebecca Viazzo Winegar
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Gerald Maxwell Cherf
- Department of Bioengineering, Stanford University, Stanford CA 94305
- Current Address: Denali Therapeutics, South San Francisco, CA, USA
| | - Dean Slocum
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - P. Daniel Poulson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Garrett E. Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | - Jonard Corpuz Valdoz
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Marcos C. Miranda
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Current Address: Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hua Bai
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Yakov Kipnis
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Audrey Olshefsky
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Tanu Priya
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Current Address: Department of Pharmacology, Northwestern University Feinberg School of Medicine; Chicago, IL 60611, USA
| | - Lauren Carter
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Cameron M. Chow
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Max R. Johnson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Suna Cheng
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - McKaela Smith
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Catherine Overed-Sayer
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K. Finch
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Alchemab Therapeutics Ltd, Cambridge, United Kingdom
| | - David Lowe
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Evox Therapeutics Limited, Oxford Science Park, Medawar Centre, East Building, Robert Robinson Avenue, Oxford, OX4 4HG
| | - Asim K. Bera
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington
| | - Timothy P Birkland
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Frank DiMaio
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Lance J. Stewart
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Melody G. Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Pam M. Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Godwin ARF, Dajani R, Zhang X, Thomson J, Holmes DF, Adamo CS, Sengle G, Sherratt MJ, Roseman AM, Baldock C. Fibrillin microfibril structure identifies long-range effects of inherited pathogenic mutations affecting a key regulatory latent TGFβ-binding site. Nat Struct Mol Biol 2023; 30:608-618. [PMID: 37081316 DOI: 10.1038/s41594-023-00950-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/28/2023] [Indexed: 04/22/2023]
Abstract
Genetic mutations in fibrillin microfibrils cause serious inherited diseases, such as Marfan syndrome and Weill-Marchesani syndrome (WMS). These diseases typically show major dysregulation of tissue development and growth, particularly in skeletal long bones, but links between the mutations and the diseases are unknown. Here we describe a detailed structural analysis of native fibrillin microfibrils from mammalian tissue by cryogenic electron microscopy. The major bead region showed pseudo eightfold symmetry where the amino and carboxy termini reside. On the basis of this structure, we show that a WMS deletion mutation leads to the induction of a structural rearrangement that blocks interaction with latent TGFβ-binding protein-1 at a remote site. Separate deletion of this binding site resulted in the assembly of shorter fibrillin microfibrils with structural alterations. The integrin αvβ3-binding site was also mapped onto the microfibril structure. These results establish that in complex extracellular assemblies, such as fibrillin microfibrils, mutations may have long-range structural consequences leading to the disruption of growth factor signaling and the development of disease.
Collapse
Affiliation(s)
- Alan R F Godwin
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rana Dajani
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Xinyang Zhang
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jennifer Thomson
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David F Holmes
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Christin S Adamo
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics, Cologne, Germany
| | - Michael J Sherratt
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Alan M Roseman
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Clair Baldock
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
30
|
Bachmann M, Kessler J, Burri E, Wehrle-Haller B. New tools to study the interaction between integrins and latent TGFβ1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525682. [PMID: 36747767 PMCID: PMC9901185 DOI: 10.1101/2023.01.26.525682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Transforming growth factor beta (TGFβ) 1 regulates cell differentiation and proliferation in different physiological settings, but is also involved in fibrotic progression and protects tumors from the immune system. Integrin αVβ6 has been shown to activate latent TGFβ1 by applying mechanical forces onto the latency-associated peptide (LAP). While the extracellular binding between αVβ6 and LAP1 is well characterized, less is known about the cytoplasmic adaptations that enable αVβ6 to apply such forces. Here, we generated new tools to facilitate the analysis of this interaction. We combined the integrin-binding part of LAP1 with a GFP and the Fc chain of human IgG. This chimeric protein, sLAP1, revealed a mechanical rearrangement of immobilized sLAP1 by αVβ6 integrin. This unique interaction was not observed between sLAP1 and other integrins. We also analyzed αVβ6 integrin binding to LAP2 and LAP3 by creating respective sLAPs. Compared to sLAP1, integrin αVβ6 showed less binding to sLAP3 and no rearrangement. These observations indicate differences in the binding of αVβ6 to LAP1 and LAP3 that have not been appreciated so far. Finally, αVβ6-sLAP1 interaction was maintained even at strongly reduced cellular contractility, highlighting the special mechanical connection between αVβ6 integrin and latent TGFβ1.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Geneva, Switzerland
| | - Jérémy Kessler
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Geneva, Switzerland
| | - Elisa Burri
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Geneva, Switzerland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Geneva, Switzerland
| |
Collapse
|
31
|
Tvaroška I, Kozmon S, Kóňa J. Molecular Modeling Insights into the Structure and Behavior of Integrins: A Review. Cells 2023; 12:cells12020324. [PMID: 36672259 PMCID: PMC9856412 DOI: 10.3390/cells12020324] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Integrins are heterodimeric glycoproteins crucial to the physiology and pathology of many biological functions. As adhesion molecules, they mediate immune cell trafficking, migration, and immunological synapse formation during inflammation and cancer. The recognition of the vital roles of integrins in various diseases revealed their therapeutic potential. Despite the great effort in the last thirty years, up to now, only seven integrin-based drugs have entered the market. Recent progress in deciphering integrin functions, signaling, and interactions with ligands, along with advancement in rational drug design strategies, provide an opportunity to exploit their therapeutic potential and discover novel agents. This review will discuss the molecular modeling methods used in determining integrins' dynamic properties and in providing information toward understanding their properties and function at the atomic level. Then, we will survey the relevant contributions and the current understanding of integrin structure, activation, the binding of essential ligands, and the role of molecular modeling methods in the rational design of antagonists. We will emphasize the role played by molecular modeling methods in progress in these areas and the designing of integrin antagonists.
Collapse
Affiliation(s)
- Igor Tvaroška
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Correspondence:
| | - Stanislav Kozmon
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| | - Juraj Kóňa
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| |
Collapse
|
32
|
Corti A, Anderluzzi G, Curnis F. Neuropilin-1 and Integrins as Receptors for Chromogranin A-Derived Peptides. Pharmaceutics 2022; 14:2555. [PMID: 36559048 PMCID: PMC9785887 DOI: 10.3390/pharmaceutics14122555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Human chromogranin A (CgA), a 439 residue-long member of the "granin" secretory protein family, is the precursor of several peptides and polypeptides involved in the regulation of the innate immunity, cardiovascular system, metabolism, angiogenesis, tissue repair, and tumor growth. Despite the many biological activities observed in experimental and preclinical models for CgA and its most investigated fragments (vasostatin-I and catestatin), limited information is available on the receptor mechanisms underlying these effects. The interaction of vasostatin-1 with membrane phospholipids and the binding of catestatin to nicotinic and b2-adrenergic receptors have been proposed as important mechanisms for some of their effects on the cardiovascular and sympathoadrenal systems. Recent studies have shown that neuropilin-1 and certain integrins may also work as high-affinity receptors for CgA, vasostatin-1 and other fragments. In this case, we review the results of these studies and discuss the structural requirements for the interactions of CgA-related peptides with neuropilin-1 and integrins, their biological effects, their mechanisms, and the potential exploitation of compounds that target these ligand-receptor systems for cancer diagnosis and therapy. The results obtained so far suggest that integrins (particularly the integrin avb6) and neuropilin-1 are important receptors that mediate relevant pathophysiological functions of CgA and CgA fragments in angiogenesis, wound healing, and tumor growth, and that these interactions may represent important targets for cancer imaging and therapy.
Collapse
Affiliation(s)
- Angelo Corti
- Faculty of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Tumor Biology and Vascular Targeting Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Anderluzzi
- Tumor Biology and Vascular Targeting Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Flavio Curnis
- Tumor Biology and Vascular Targeting Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
33
|
Integrin Alpha v Beta 6 (αvβ6) and Its Implications in Cancer Treatment. Int J Mol Sci 2022; 23:ijms232012346. [PMID: 36293202 PMCID: PMC9603893 DOI: 10.3390/ijms232012346] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/20/2022] Open
Abstract
Integrins are necessary for cell adhesion, migration, and positioning. Essential for inducing signalling events for cell survival, proliferation, and differentiation, they also trigger a variety of signal transduction pathways involved in mediating invasion, metastasis, and squamous-cell carcinoma. Several recent studies have demonstrated that the up- and down-regulation of the expression of αv and other integrins can be a potent marker of malignant diseases and patient prognosis. This review focuses on an arginine-glycine-aspartic acid (RGD)-dependent integrin αVβ6, its biology, and its role in healthy humans. We examine the implications of αVβ6 in cancer progression and the promotion of epithelial-mesenchymal transition (EMT) by contributing to the activation of transforming growth factor beta TGF-β. Although αvβ6 is crucial for proper function in healthy people, it has also been validated as a target for cancer treatment. This review briefly considers aspects of targeting αVβ6 in the clinic via different therapeutic modalities.
Collapse
|
34
|
Lin FY, Li J, Xie Y, Zhu J, Huong Nguyen TT, Zhang Y, Zhu J, Springer TA. A general chemical principle for creating closure-stabilizing integrin inhibitors. Cell 2022; 185:3533-3550.e27. [PMID: 36113427 PMCID: PMC9494814 DOI: 10.1016/j.cell.2022.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/07/2022] [Accepted: 08/04/2022] [Indexed: 01/26/2023]
Abstract
Integrins are validated drug targets with six approved therapeutics. However, small-molecule inhibitors to three integrins failed in late-stage clinical trials for chronic indications. Such unfavorable outcomes may in part be caused by partial agonism, i.e., the stabilization of the high-affinity, extended-open integrin conformation. Here, we show that the failed, small-molecule inhibitors of integrins αIIbβ3 and α4β1 stabilize the high-affinity conformation. Furthermore, we discovered a simple chemical feature present in multiple αIIbβ3 antagonists that stabilizes integrins in their bent-closed conformation. Closing inhibitors contain a polar nitrogen atom that stabilizes, via hydrogen bonds, a water molecule that intervenes between a serine residue and the metal in the metal-ion-dependent adhesion site (MIDAS). Expulsion of this water is a requisite for transition to the open conformation. This change in metal coordination is general to integrins, suggesting broad applicability of the drug-design principle to the integrin family, as validated with a distantly related integrin, α4β1.
Collapse
Affiliation(s)
- Fu-Yang Lin
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Li
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yonghua Xie
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, PRC
| | - Jianghai Zhu
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Thi Thu Huong Nguyen
- Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, PRC.
| | - Jieqing Zhu
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Anti-TGF-β1 aptamer enhances therapeutic effect of tyrosine kinase inhibitor, gefitinib, on non-small cell lung cancer in xenograft model. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:969-978. [PMID: 36189081 PMCID: PMC9481871 DOI: 10.1016/j.omtn.2022.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 06/07/2022] [Indexed: 11/30/2022]
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine that plays crucial pathophysiological roles in various diseases, such as cancer and fibrosis. However, the disease modulation by targeting TGF-β1 isoform remains to be established, regardless of several studies employed with limited antibodies. Here, we developed an RNA aptamer to human active TGF-β1, named APT-β1, and characterized its properties in vitro and in vivo. APT-β1 bound to human and mouse active TGF-β1 proteins with high affinity and specificity and strongly inhibited TGF-β1-induced downstream signaling and cell morphology with 50% inhibition concentration (IC50) values at picomolar concentrations. In a xenograft mouse model of non-small cell lung cancer, APT-β1 alone showed no appreciable effect on tumor growth, while it greatly enhanced the anti-tumor effect of gefitinib, an approved tyrosine kinase inhibitor. These findings strongly suggest that the anti-TGF-β1 medication may be a promising cancer therapy to suppress repopulation of lung cancer in combination with certain anti-cancer drugs, such as gefitinib.
Collapse
|
36
|
Floresta G, Memdouh S, Pham T, Ma MT, Blower PJ, Hider RC, Abbate V, Cilibrizzi A. Targeting integrin αvβ6 with gallium-68 tris (hydroxypyridinone) based PET probes. Dalton Trans 2022; 51:12796-12803. [PMID: 35972045 PMCID: PMC9425781 DOI: 10.1039/d2dt00980c] [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: 03/30/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022]
Abstract
Expression of the cellular transmembrane receptor αvβ6 integrin is mostly restricted to malignant epithelial cells in a wide variety of carcinomas, including pancreatic and others derived from epithelial tissues. Thus, this protein is considered an attractive target for tumour imaging and therapy. Two different 68Ga hexadentate tris (3,4-hydroxypyridinone) (THP) chelators were produced in this study and coupled to the αvβ6 integrin-selective peptide cyclo(FRGDLAFp(NMe)K) via NHS chemistry. Radiolabelling experiments confirmed a high radiochemical yield of the two PET probes. In addition, cellular binding studies showed high binding affinities in the nanomolar range. The two integrin αvβ6-peptide-THP synthesized and radiolabeled in this study will facilitate in vivo monitoring of transmembrane receptor αvβ6 integrin by using the advantage of THP chemistry for rapid, efficient and stable gallium chelation.
Collapse
Affiliation(s)
- Giuseppe Floresta
- King's College London, Institute of Pharmaceutical Science, Franklin Wilkins Building, London SE1 9NH, UK.
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Siham Memdouh
- King's College London, Institute of Pharmaceutical Science, Franklin Wilkins Building, London SE1 9NH, UK.
| | - Truc Pham
- King's College London, Division of Imaging Sciences and Biomedical Engineering, Fourth Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK
| | - Michelle T Ma
- King's College London, Division of Imaging Sciences and Biomedical Engineering, Fourth Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK
| | - Philip J Blower
- King's College London, Division of Imaging Sciences and Biomedical Engineering, Fourth Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK
| | - Robert C Hider
- King's College London, Institute of Pharmaceutical Science, Franklin Wilkins Building, London SE1 9NH, UK.
| | - Vincenzo Abbate
- King's College London, Institute of Pharmaceutical Science, Franklin Wilkins Building, London SE1 9NH, UK.
| | - Agostino Cilibrizzi
- King's College London, Institute of Pharmaceutical Science, Franklin Wilkins Building, London SE1 9NH, UK.
- Centre for Therapeutic Innovation, University of Bath, Bath, UK
| |
Collapse
|
37
|
Duan Z, Lin X, Wang L, Zhen Q, Jiang Y, Chen C, Yang J, Lee CH, Qin Y, Li Y, Zhao B, Wang J, Zhang Z. Specificity of TGF-β1 signal designated by LRRC33 and integrin α Vβ 8. Nat Commun 2022; 13:4988. [PMID: 36008481 PMCID: PMC9411592 DOI: 10.1038/s41467-022-32655-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/03/2022] [Indexed: 12/20/2022] Open
Abstract
Myeloid lineage cells present the latent form of transforming growth factor-β1 (L-TGF-β1) to the membrane using an anchor protein LRRC33. Integrin αVβ8 activates extracellular L-TGF-β1 to trigger the downstream signaling functions. However, the mechanism designating the specificity of TGF-β1 presentation and activation remains incompletely understood. Here, we report cryo-EM structures of human L-TGF-β1/LRRC33 and integrin αVβ8/L-TGF-β1 complexes. Combined with biochemical and cell-based analyses, we demonstrate that LRRC33 only presents L-TGF-β1 but not the -β2 or -β3 isoforms due to difference of key residues on the growth factor domains. Moreover, we reveal a 2:2 binding mode of integrin αVβ8 and L-TGF-β1, which shows higher avidity and more efficient L-TGF-β1 activation than previously reported 1:2 binding mode. We also uncover that the disulfide-linked loop of the integrin subunit β8 determines its exquisite affinity to L-TGF-β1. Together, our findings provide important insights into the specificity of TGF-β1 signaling achieved by LRRC33 and integrin αVβ8.
Collapse
Affiliation(s)
- Zelin Duan
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Xuezhen Lin
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Lixia Wang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Qiuxin Zhen
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Yuefeng Jiang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Chuxin Chen
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yan Qin
- Parthenon Therapeutics, 40 Guest street, Boston, MA, 02135, USA
| | - Ying Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Bo Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China.
| | - Jianchuan Wang
- Center for Translational Research, Shenzhen Bay Laboratory, 518007, Shenzhen, Guangdong, China.
| | - Zhe Zhang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China.
| |
Collapse
|
38
|
Heterotropic roles of divalent cations in the establishment of allostery and affinity maturation of integrin αXβ2. Cell Rep 2022; 40:111254. [PMID: 36001965 PMCID: PMC9440770 DOI: 10.1016/j.celrep.2022.111254] [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: 06/17/2021] [Revised: 05/23/2022] [Accepted: 08/02/2022] [Indexed: 11/21/2022] Open
Abstract
Allosteric activation and silencing of leukocyte β2-integrins transpire through cation-dependent structural changes, which mediate integrin biosynthesis and recycling, and are essential to designing leukocyte-specific drugs. Stepwise addition of Mg2+ reveals two mutually coupled events for the αXβ2 ligand-binding domain-the αX I-domain-corresponding to allostery establishment and affinity maturation. Electrostatic alterations in the Mg2+-binding site establish long-range couplings, leading to both pH- and Mg2+-occupancy-dependent biphasic stability change in the αX I-domain fold. The ligand-binding sensorgrams show composite affinity events for the αX I-domain accounting for the multiplicity of the αX I-domain conformational states existing in the solution. On cell surfaces, increasing Mg2+ concentration enhanced adhesiveness of αXβ2. This work highlights how intrinsically flexible pH- and cation-sensitive architecture endows a unique dynamic continuum to the αI-domain structure on the intact integrin, thereby revealing the importance of allostery establishment and affinity maturation in both extracellular and intracellular integrin events.
Collapse
|
39
|
Bugatti K, Andreucci E, Monaco N, Battistini L, Peppicelli S, Ruzzolini J, Curti C, Zanardi F, Bianchini F, Sartori A. Nintedanib-Containing Dual Conjugates Targeting α Vβ 6 Integrin and Tyrosine Kinase Receptors as Potential Antifibrotic Agents. ACS OMEGA 2022; 7:17658-17669. [PMID: 35664627 PMCID: PMC9161413 DOI: 10.1021/acsomega.2c00535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 06/15/2023]
Abstract
αVβ6 Integrin plays a fundamental role in the activation of transforming growth factor-β (TGF-β), the major profibrotic mediator; for this reason, αVβ6 ligands have recently been forwarded to clinical phases for the therapy of fibrotic diseases. Herein, we report the synthesis and in vitro biological evaluation as antifibrotic agents of three new covalent conjugates, constituted by c(AmpLRGDL), an αVβ6 integrin-recognizing small cyclopeptide, and nintedanib, a tyrosine kinase inhibitor approved for idiopathic pulmonary fibrosis (IPF) treatment. One of these conjugates recapitulates optimal in vitro antifibrotic properties of the two active units. The integrin ligand portion within the conjugate plays a role in inhibiting profibrotic stimuli, potentiating the nintedanib effect and favoring the selective uptake of the conjugate in cells overexpressing αVβ6 integrin. These results may open a new perspective on the development of dual conjugates in the targeted therapy of IPF.
Collapse
Affiliation(s)
- Kelly Bugatti
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Elena Andreucci
- Department
of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Noemi Monaco
- Department
of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Lucia Battistini
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Silvia Peppicelli
- Department
of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Jessica Ruzzolini
- Department
of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Claudio Curti
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Franca Zanardi
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Francesca Bianchini
- Department
of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Andrea Sartori
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27A, 43124 Parma, Italy
| |
Collapse
|
40
|
Chen SY, Mamai O, Akhurst RJ. TGFβ: Signaling Blockade for Cancer Immunotherapy. ANNUAL REVIEW OF CANCER BIOLOGY 2022; 6:123-146. [PMID: 36382146 PMCID: PMC9645596 DOI: 10.1146/annurev-cancerbio-070620-103554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Discovered over four decades ago, transforming growth factor β (TGFβ) is a potent pleiotropic cytokine that has context-dependent effects on most cell types. It acts as a tumor suppressor in some cancers and/or supports tumor progression and metastasis through its effects on the tumor stroma and immune microenvironment. In TGFβ-responsive tumors it can promote invasion and metastasis through epithelial-mesenchymal transformation, the appearance of cancer stem cell features, and resistance to many drug classes, including checkpoint blockade immunotherapies. Here we consider the biological activities of TGFβ action on different cells of relevance toward improving immunotherapy outcomes for patients, with a focus on the adaptive immune system. We discuss recent advances in the development of drugs that target the TGFβ signaling pathway in a tumor-specific or cell type–specific manner to improve the therapeutic window between response rates and adverse effects.
Collapse
Affiliation(s)
- Szu-Ying Chen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Ons Mamai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Rosemary J. Akhurst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
- Department of Anatomy, University of California, San Francisco, California, USA
| |
Collapse
|
41
|
De A, Morales JE, Chen Z, Sebastian S, McCarty JH. The β8 integrin cytoplasmic domain activates extracellular matrix adhesion to promote brain neurovascular development. Development 2022; 149:274538. [PMID: 35217866 PMCID: PMC8977100 DOI: 10.1242/dev.200472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/11/2022] [Indexed: 12/11/2022]
Abstract
In the developing mammalian brain, neuroepithelial cells interact with blood vessels to regulate angiogenesis, blood-brain barrier maturation and other key neurovascular functions. Genetic studies in mice have shown that neurovascular development is controlled, in part, by Itgb8, which encodes the neuroepithelial cell-expressed integrin β8 subunit. However, these studies have involved complete loss-of-function Itgb8 mutations, and have not discerned the relative roles for the β8 integrin extracellular matrix (ECM) binding region versus the intracellular signaling tail. Here, Cre/lox strategies have been employed to selectively delete the cytoplasmic tail of murine Itgb8 without perturbing its transmembrane and extracellular domains. We report that the β8 integrin cytoplasmic domain is essential for inside-out modulation of adhesion, including activation of latent-TGFβs in the ECM. Quantitative sequencing of the brain endothelial cell transcriptome identifies TGFβ-regulated genes with putative links to blood vessel morphogenesis, including several genes linked to Wnt/β-catenin signaling. These results reveal that the β8 integrin cytoplasmic domain is essential for the regulation of TGFβ-dependent gene expression in endothelial cells and suggest that cross-talk between TGFβs and Wnt pathways is crucial for neurovascular development.
Collapse
Affiliation(s)
- Arpan De
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - John E Morales
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Zhihua Chen
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Sumod Sebastian
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Joseph H McCarty
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|
42
|
Abstract
Transforming growth factor-β (TGFβ) signalling controls multiple cell fate decisions during development and tissue homeostasis; hence, dysregulation of this pathway can drive several diseases, including cancer. Here we discuss the influence that TGFβ exerts on the composition and behaviour of different cell populations present in the tumour immune microenvironment, and the context-dependent functions of this cytokine in suppressing or promoting cancer. During homeostasis, TGFβ controls inflammatory responses triggered by exposure to the outside milieu in barrier tissues. Lack of TGFβ exacerbates inflammation, leading to tissue damage and cellular transformation. In contrast, as tumours progress, they leverage TGFβ to drive an unrestrained wound-healing programme in cancer-associated fibroblasts, as well as to suppress the adaptive immune system and the innate immune system. In consonance with this key role in reprogramming the tumour microenvironment, emerging data demonstrate that TGFβ-inhibitory therapies can restore cancer immunity. Indeed, this approach can synergize with other immunotherapies - including immune checkpoint blockade - to unleash robust antitumour immune responses in preclinical cancer models. Despite initial challenges in clinical translation, these findings have sparked the development of multiple therapeutic strategies that inhibit the TGFβ pathway, many of which are currently in clinical evaluation.
Collapse
Affiliation(s)
- Daniele V F Tauriello
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
43
|
Bieri M, Hendrickx R, Bauer M, Yu B, Jetzer T, Dreier B, Mittl PRE, Sobek J, Plückthun A, Greber UF, Hemmi S. The RGD-binding integrins αvβ6 and αvβ8 are receptors for mouse adenovirus-1 and -3 infection. PLoS Pathog 2021; 17:e1010083. [PMID: 34910784 PMCID: PMC8673666 DOI: 10.1371/journal.ppat.1010083] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Mammalian adenoviruses (AdVs) comprise more than ~350 types including over 100 human (HAdVs) and just three mouse AdVs (MAdVs). While most HAdVs initiate infection by high affinity/avidity binding of their fiber knob (FK) protein to either coxsackievirus AdV receptor (CAR), CD46 or desmoglein (DSG)-2, MAdV-1 (M1) infection requires arginine-glycine-aspartate (RGD) binding integrins. To identify the receptors mediating MAdV infection we generated five novel reporter viruses for MAdV-1/-2/-3 (M1, M2, M3) transducing permissive murine (m) CMT-93 cells, but not B16 mouse melanoma cells expressing mCAR, human (h) CD46 or hDSG-2. Recombinant M1 or M3 FKs cross-blocked M1 and M3 but not M2 infections. Profiling of murine and human cells expressing RGD-binding integrins suggested that αvβ6 and αvβ8 heterodimers are associated with M1 and M3 infections. Ectopic expression of mβ6 in B16 cells strongly enhanced M1 and M3 binding, infection, and progeny production comparable with mαvβ6-positive CMT-93 cells, whereas mβ8 expressing cells were more permissive to M1 than M3. Anti-integrin antibodies potently blocked M1 and M3 binding and infection of CMT-93 cells and hαvβ8-positive M000216 cells. Soluble integrin αvβ6, and synthetic peptides containing the RGDLXXL sequence derived from FK-M1, FK-M3 and foot and mouth disease virus coat protein strongly interfered with M1/M3 infections, in agreement with high affinity interactions of FK-M1/FK-M3 with αvβ6/αvβ8, determined by surface plasmon resonance measurements. Molecular docking simulations of ternary complexes revealed a bent conformation of RGDLXXL-containing FK-M3 peptides on the subunit interface of αvβ6/β8, where the distal leucine residue dips into a hydrophobic pocket of β6/8, the arginine residue ionically engages αv aspartate215, and the aspartate residue coordinates a divalent cation in αvβ6/β8. Together, the RGDLXXL-bearing FKs are part of an essential mechanism for M1/M3 infection engaging murine and human αvβ6/8 integrins. These integrins are highly conserved in other mammals, and may favour cross-species virus transmission.
Collapse
Affiliation(s)
- Manuela Bieri
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University Of Zurich, Switzerland
| | - Rodinde Hendrickx
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University Of Zurich, Switzerland
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tania Jetzer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Peer R. E. Mittl
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule (ETH) Zurich and University of Zurich, Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Silvio Hemmi
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Yokosaki Y, Nishimichi N. New Therapeutic Targets for Hepatic Fibrosis in the Integrin Family, α8β1 and α11β1, Induced Specifically on Activated Stellate Cells. Int J Mol Sci 2021; 22:12794. [PMID: 34884600 PMCID: PMC8657911 DOI: 10.3390/ijms222312794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
A huge effort has been devoted to developing drugs targeting integrins over 30 years, because of the primary roles of integrins in the cell-matrix milieu. Five αv-containing integrins, in the 24 family members, have been a central target of fibrosis. Currently, a small molecule against αvβ1 is undergoing a clinical trial for NASH-associated fibrosis as a rare agent aiming at fibrogenesis. Latent TGFβ activation, a distinct talent of αv-integrins, has been intriguing as a therapeutic target. None of the αv-integrin inhibitors, however, has been in the clinical market. αv-integrins commonly recognize an Arg-Gly-Asp (RGD) sequence, and thus the pharmacophore of inhibitors for the 5-integrins is based on the same RGD structure. The RGD preference of the integrins, at the same time, dilutes ligand specificity, as the 5-integrins share ligands containing RGD sequence such as fibronectin. With the inherent little specificity in both drugs and targets, "disease specificity" has become less important for the inhibitors than blocking as many αv-integrins. In fact, an almighty inhibitor for αv-integrins, pan-αv, was in a clinical trial. On the contrary, approved integrin inhibitors are all specific to target integrins, which are expressed in a cell-type specific manner: αIIbβ3 on platelets, α4β1, α4β7 and αLβ2 on leukocytes. Herein, "disease specific" integrins would serve as attractive targets. α8β1 and α11β1 are selectively expressed in hepatic stellate cells (HSCs) and distinctively induced upon culture activation. The exceptional specificity to activated HSCs reflects a rather "pathology specific" nature of these new integrins. The monoclonal antibodies against α8β1 and α11β1 in preclinical examinations may illuminate the road to the first medical agents.
Collapse
Affiliation(s)
- Yasuyuki Yokosaki
- Integrin-Matrix Biomedical Science, Translational Research Center, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan;
| | | |
Collapse
|
45
|
Garlatti V, Lovisa S, Danese S, Vetrano S. The Multiple Faces of Integrin-ECM Interactions in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:10439. [PMID: 34638778 PMCID: PMC8508809 DOI: 10.3390/ijms221910439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) comprises a series of chronic and relapsing intestinal diseases, with Crohn's disease and ulcerative colitis being the most common. The abundant and uncontrolled deposition of extracellular matrix, namely fibrosis, is one of the major hallmarks of IBD and is responsible for the progressive narrowing and closure of the intestine, defined as stenosis. Although fibrosis is usually considered the product of chronic inflammation, the substantial failure of anti-inflammatory therapies to target and reduce fibrosis in IBD suggests that fibrosis might be sustained in an inflammation-independent manner. Pharmacological therapies targeting integrins have recently shown great promise in the treatment of IBD. The efficacy of these therapies mainly relies on their capacity to target the integrin-mediated recruitment and functionality of the immune cells at the damage site. However, by nature, integrins also act as mechanosensitive molecules involved in the intracellular transduction of signals and modifications originating from the extracellular matrix. Therefore, understanding integrin signaling in the context of IBD may offer important insights into mechanisms of matrix remodeling, which are uncoupled from inflammation and could underlie the onset and persistency of intestinal fibrosis. In this review, we present the currently available knowledge on the role of integrins in the etiopathogenesis of IBD, highlighting their role in the context of immune-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Valentina Garlatti
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale ‘A. Avogadro’, 28100 Novara, Italy
| | - Sara Lovisa
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Silvio Danese
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Stefania Vetrano
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
46
|
Arimori T, Miyazaki N, Mihara E, Takizawa M, Taniguchi Y, Cabañas C, Sekiguchi K, Takagi J. Structural mechanism of laminin recognition by integrin. Nat Commun 2021; 12:4012. [PMID: 34188035 PMCID: PMC8241838 DOI: 10.1038/s41467-021-24184-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Recognition of laminin by integrin receptors is central to the epithelial cell adhesion to basement membrane, but the structural background of this molecular interaction remained elusive. Here, we report the structures of the prototypic laminin receptor α6β1 integrin alone and in complex with three-chain laminin-511 fragment determined via crystallography and cryo-electron microscopy, respectively. The laminin-integrin interface is made up of several binding sites located on all five subunits, with the laminin γ1 chain C-terminal portion providing focal interaction using two carboxylate anchor points to bridge metal-ion dependent adhesion site of integrin β1 subunit and Asn189 of integrin α6 subunit. Laminin α5 chain also contributes to the affinity and specificity by making electrostatic interactions with large surface on the β-propeller domain of α6, part of which comprises an alternatively spliced X1 region. The propeller sheet corresponding to this region shows unusually high mobility, suggesting its unique role in ligand capture.
Collapse
Affiliation(s)
- Takao Arimori
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Naoyuki Miyazaki
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan ,grid.20515.330000 0001 2369 4728Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Emiko Mihara
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Mamoru Takizawa
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Yukimasa Taniguchi
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Carlos Cabañas
- grid.465524.4Cell-cell Communication & Inflammation Unit, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Immunology, Ophthalmology and Otorhinolaryngology (IOO), Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain ,grid.144756.50000 0001 1945 5329Instituto de Investigación Sanitaria Hospital 12 Octubre (i+12), Madrid, Spain
| | - Kiyotoshi Sekiguchi
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Junichi Takagi
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| |
Collapse
|
47
|
Inhibition of Cancer Cell Adhesion, Migration and Proliferation by a Bispecific Antibody that Targets two Distinct Epitopes on αv Integrins. J Mol Biol 2021; 433:167090. [PMID: 34090922 DOI: 10.1016/j.jmb.2021.167090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 01/13/2023]
Abstract
Members of the αv family of integrins regulate activation of transforming growth factor beta (TGFβ) and are directly involved in pro-tumorigenic phenotypes. Thus, αv integrins may be therapeutic targets for fibrosis and cancer, yet the isolation of selective inhibitors is currently a challenge. We generated synthetic antibodies selective for αv integrins by phage display selections on cell lines that displayed integrin heterodimers. We identified antibodies that targeted two distinct epitopes on cell-surface αv integrins and partially inhibited cell adhesion mediated by interactions between integrins and the latency-associated peptide, part of the pro-form of TGFβ. Using the isolated antibody paratope sequences we engineered a bispecific antibody capable of binding to both epitopes simultaneously; this antibody potently and completely inhibited cell adhesion mediated by integrins αvβ1, αvβ3 and αvβ5. In addition, the bispecific antibody inhibited proliferation and migration of lung carcinoma lines, where the highest and lowest potencies observed correlated with integrin-αv cell surface expression levels. Taken together, our results demonstrate that phage display selections with live cells can yield high quality anti-integrin antibodies, which we used as biparatopic building blocks to construct a bispecific antibody that strongly inhibited integrin function and may be a therapeutic candidate for cancer and fibrosis.
Collapse
|
48
|
Zhen G, Guo Q, Li Y, Wu C, Zhu S, Wang R, Guo XE, Kim BC, Huang J, Hu Y, Dan Y, Wan M, Ha T, An S, Cao X. Mechanical stress determines the configuration of TGFβ activation in articular cartilage. Nat Commun 2021; 12:1706. [PMID: 33731712 PMCID: PMC7969741 DOI: 10.1038/s41467-021-21948-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
Our incomplete understanding of osteoarthritis (OA) pathogenesis has significantly hindered the development of disease-modifying therapy. The functional relationship between subchondral bone (SB) and articular cartilage (AC) is unclear. Here, we found that the changes of SB architecture altered the distribution of mechanical stress on AC. Importantly, the latter is well aligned with the pattern of transforming growth factor beta (TGFβ) activity in AC, which is essential in the regulation of AC homeostasis. Specifically, TGFβ activity is concentrated in the areas of AC with high mechanical stress. A high level of TGFβ disrupts the cartilage homeostasis and impairs the metabolic activity of chondrocytes. Mechanical stress stimulates talin-centered cytoskeletal reorganization and the consequent increase of cell contractile forces and cell stiffness of chondrocytes, which triggers αV integrin-mediated TGFβ activation. Knockout of αV integrin in chondrocytes reversed the alteration of TGFβ activation and subsequent metabolic abnormalities in AC and attenuated cartilage degeneration in an OA mouse model. Thus, SB structure determines the patterns of mechanical stress and the configuration of TGFβ activation in AC, which subsequently regulates chondrocyte metabolism and AC homeostasis.
Collapse
Affiliation(s)
- Gehua Zhen
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Yusheng Li
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Chuanlong Wu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Shouan Zhu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Ruomei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Byoung Choul Kim
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Jessie Huang
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Dan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Steven An
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
49
|
Possible Action of Transition Divalent Metal Ions at the Inter-Pentameric Interface of Inactivated Foot-and-Mouth Disease Virus Provide A Simple but Effective Approach to Enhance Stability. J Virol 2021; 95:JVI.02431-20. [PMID: 33441340 PMCID: PMC8092711 DOI: 10.1128/jvi.02431-20] [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] [Indexed: 12/13/2022] Open
Abstract
The structural instability of inactivated foot-and-mouth disease virus (FMDV) hinders the development of vaccine industry. Here we found that some transition metal ions like Cu2+ and Ni2+ could specifically bind to FMDV capsids at capacities about 7089 and 3448 metal ions per capsid, respectively. These values are about 33- and 16-folds of the binding capacity of non-transition metal ion Ca2+ (about 214 per capsid). Further thermodynamic studies indicated that all these three metal ions bound to the capsids in spontaneous enthalpy driving manners (ΔG<0, ΔH<0, ΔS<0), and the Cu2+ binding had the highest affinity. The binding of Cu2+ and Ni2+ could enhance both the thermostability and acid-resistant stability of capsids, while the binding of Ca2+ was helpful only to the thermostability of the capsids. Animal experiments showed that the immunization of FMDV bound with Cu2+ induced the highest specific antibody titers in mice. Coincidently, the FMDV bound with Cu2+ exhibited significantly enhanced affinities to integrin β6 and heparin sulfate, both of which are important cell surface receptors for FMDV attaching. Finally, the specific interaction between capsids and Cu2+ or Ni2+ was applied to direct purification of FMDV from crude cell culture feedstock by the immobilized metal affinity chromatography. Based on our new findings and structural analysis of the FMDV capsid, a "transition metal ion bridges" mechanism that describes linkage between adjacent histidine and other amino acids at the inter-pentameric interface of the capsids by transition metal ions coordination action was proposed to explain their stabilizing effect imposed on the capsid.IMPORTANCE How to stabilize the inactivated FMDV without affecting virus infectivity and immunogenicity is a big challenge in vaccine industry. The electrostatic repulsion induced by protonation of a large amount of histidine residues at the inter-pentameric interface of viral capsids is one of the major mechanisms causing the dissociation of capsids. In the present work, this structural disadvantage inspired us to stabilize the capsids through coordinating transition metal ions with the adjacent histidine residues in FMDV capsid, instead of removing or substituting them. This approach was proved effective to enhance not only the stability of FMDV, but also enhance the specific antibody responses; thus, providing a new guideline for designing an easy-to-use strategy suitable for large-scale production of FMDV vaccine antigen.
Collapse
|
50
|
Zhang J, Ten Dijke P, Wuhrer M, Zhang T. Role of glycosylation in TGF-β signaling and epithelial-to-mesenchymal transition in cancer. Protein Cell 2021; 12:89-106. [PMID: 32583064 PMCID: PMC7862465 DOI: 10.1007/s13238-020-00741-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Glycosylation is a common posttranslational modification on membrane-associated and secreted proteins that is of pivotal importance for regulating cell functions. Aberrant glycosylation can lead to uncontrolled cell proliferation, cell-matrix interactions, migration and differentiation, and has been shown to be involved in cancer and other diseases. The epithelial-to-mesenchymal transition is a key step in the metastatic process by which cancer cells gain the ability to invade tissues and extravasate into the bloodstream. This cellular transformation process, which is associated by morphological change, loss of epithelial traits and gain of mesenchymal markers, is triggered by the secreted cytokine transforming growth factor-β (TGF-β). TGF-β bioactivity is carefully regulated, and its effects on cells are mediated by its receptors on the cell surface. In this review, we first provide a brief overview of major types of glycans, namely, N-glycans, O-glycans, glycosphingolipids and glycosaminoglycans that are involved in cancer progression. Thereafter, we summarize studies on how the glycosylation of TGF-β signaling components regulates TGF-β secretion, bioavailability and TGF-β receptor function. Then, we review glycosylation changes associated with TGF-β-induced epithelial-to-mesenchymal transition in cancer. Identifying and understanding the mechanisms by which glycosylation affects TGF-β signaling and downstream biological responses will facilitate the identification of glycans as biomarkers and enable novel therapeutic approaches.
Collapse
Affiliation(s)
- Jing Zhang
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands.
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|