Open Access

TMEM16 proteins: Ca2+‑activated chloride channels and phospholipid scramblases as potential drug targets (Review)

  • Authors:
    • Zeqi Huang
    • Zoya Iqbal
    • Zhe Zhao
    • Xiaoqiang Chen
    • Ayesha Mahmmod
    • Jianquan Liu
    • Wencui Li
    • Zhiqin Deng
  • View Affiliations

  • Published online on: July 29, 2024     https://doi.org/10.3892/ijmm.2024.5405
  • Article Number: 81
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

TMEM16 proteins, which function as Ca2+‑activated Cl channels are involved in regulating a wide variety of cellular pathways and functions. The modulators of Cl channels can be used for the molecule‑based treatment of respiratory diseases, cystic fibrosis, tumors, cancer, osteoporosis and coronavirus disease 2019. The TMEM16 proteins link Ca2+ signaling, cellular electrical activity and lipid transport. Thus, deciphering these complex regulatory mechanisms may enable a more comprehensive understanding of the physiological functions of the TMEM16 proteins and assist in ascertaining the applicability of these proteins as potential pharmacological targets for the treatment of a range of diseases. The present review examined the structures, functions and characteristics of the different types of TMEM16 proteins, their association with the pathogenesis of various diseases and the applicability of TMEM16 modulator‑based treatment methods.

Introduction

The Cl channels present in mammalian cells can be divided into the following five categories: Cystic fibrosis transmembrane conductance regulator (CFTR), Ca2+-activated Cl channels (CaCCs), ligand-gated Cl channels, volume-regulated Cl channels and voltage-gated Cl channels (1). The TMEM16 proteins are Cl channels that can reveal one or more regulatory mechanism, ensuring the normal functioning of molecules or compounds in an organism through their protein typing (2). At present, 10 subtypes of TMEM16 proteins have been identified, which play a role in various functions of the human body (3). Certain members, such as TMEM16A and B, serve as CaCCs and as phospholipid scramblases, thereby demonstrating the dual functions of channels and scramblases, with others performing other cellular functions (4). Transfection of other TMEM16 proteins (such as 16C, 16F, 16G, 16H, 16J and 16K) into null cells does not result in increased anion transport or membrane current, indicating that these proteins have other cellular functions (4). For instance, research has shown that TMEM16J (anostatin-9) exhibits activity as a cAMP/PKA activated channel (5). The TMEM16 proteins are activated by the Gq protein-coupled receptor, mutations of which lead to cellular dysfunction (6). The TMEM16 proteins link Ca2+ signals with cellular electrical activity and lipid transport and play a critical role in the manifestation and proliferation of various human diseases, including cystic fibrosis (CF), jaw dysplasia, nephrolithiasis, myotonia congenita and cancer (7). Therefore, fully understanding the structures and functions of the TMEM16 proteins, unraveling the mechanisms underlying their involvement in complex molecular transport and ascertaining their potential as targets for the design of drugs for the treatment of human diseases are major avenues of pursued research.

Materials and methods

The present narrative review followed the Assessment of Narrative Review Articles flowchart (Fig. 1) (8). The main purpose of the present review was to summarize the evidence of potential therapeutic targets in TMEM16 protein research and to understand the basic characteristic structures, gene mutations and treatment strategies for related diseases. The English terms ['TMEM16' (Mesh)] OR ['TMEM16' (Mesh)] AND ['Structure' (Mesh)] OR ['Treatment' (Mesh)] were searched on the PubMed (https://pubmed.ncbi.nlm.nih.gov), Google Scholar (https://scholar.google.com) and Cochrane databases (https://www.cochranelibrary.com). The screening results included literature published in the past 15 years. Articles that met the following criteria were included in the present review: i) The data reported in the study was from animals; ii) the structure of TMEM16 protein could be found in the corresponding data in the Protein Data Bank (PDB, https://www.rcsb.org); iii) cases of clinical diseases were not individual case studies; iv) clearly stated the specific methods and evidence was supported by referenced citations; and v) the articles were not practical guidelines, guidelines, meta-analyses, systematic reviews, narrative reviews, case series and case reports. Articles that did not describe the methods and those that were not strictly related to the research objectives were excluded. The search strategy identified 221 articles, of which 192 were excluded after evaluating the title and abstract. Then, based on the importance of the journal, including comparison of research designs and methods, evaluation of journal papers, journal impact factors, academic reputation of scholars and academic status of institutions, three independent reviewers studied the titles and abstracts of the articles. To prioritize analysis of the various subtypes of TMEM16 protein and their corresponding cellular functions, and to analyze the disease and treatment targets based on the collected literature, 29 articles were selected for quality evaluation. The abstracts and images of all selected articles were reviewed and the data and content from the complete article were ultimately used to write the present review.

Structural characteristics of TMEM16 proteins

The TMEM16 superfamily comprises bifunctional Cl channels and phospholipid scramblases. These two isoforms possess a common homodimeric structure with the transmembrane domains (TM) 3-7 helix forming a hydrophilic groove, the multiple conformations of which allow the passage of ions and lipids (Fig. 2) (9,10). The dimer formed by the TMEM16 proteins is surrounded by TM3-7, with each subunit of the dimer possessing an ion-conducting pore (9). The Ca2+-dependent activation of TMEM16, which involves the direct binding of Ca2+, induces the formation of three consecutive Ca2+-binding sites between TM6 and 8 in each subunit (11). The TMEM16F structure, as with TMEM16A, has a large extracellular domain formed by the extracellular loops of TM1-2, 3-4, 5-6, 7-8 and 9-10, and is stabilized by four disulfide bonds. Disruption of TMEM16F leads to ion channel dysfunction (6,7). Nevertheless, the Ca2+-binding regions of TMEM16A and F can adopt a closed permeation pathway in which the pores are too narrow to allow the passage of ions (9). This closed conformation is likely due to a Ca2+-dependent run-down (desensitization) (10). The proteins belonging to the TMEM16 family are also known as anoctamins, 10 of which are 800-1,000 amino acids long and are suffixed with the letters A-K, excluding I (9). The structure of each family member comprises 10 transmembrane domains, with the -NH2 and -COOH termini inserted within the cytoplasmic matrix (9). The -NH2 terminus consists of a dimeric domain involved in homotypic interactions with TMEM16A proteins (10). Two main models have explained the structural characteristics of the TMEM16A proteins (12). The first model suggests the formation of a repetitive reentrant loop between TM5 and 6, which is important for forming the ion channels (12). A previous study also revealed that the EEEEEEAVK sequence in the first intracellular loop has an essential role in the Ca2+-dependent regulation of the voltage mechanism (13). The second model proposes a transmembrane domain that directly crosses the cell membrane into the cytoplasmic matrix to form a loop between TM6 and 7, which is important for Ca2+ as this loop forms the sixth transmembrane domain that binds to Ca2+, regulating channel activity (13). The main Ca2+ binding site of the model is located in the third intracellular loop, between two Glu residues (702 and 705) that mediate the activation of the Ca2+ channel. The fourth main intracellular loop may also harbor Ca2+ binding sites. The messenger protein, such as calmodulin, possesses three sites for Ca2+ binding: Calmodulin-binding motifs 1 and 2, which separately have calmodulin-binding activity (14), and the regulatory calmodulin binding site, which provides two different modes of interaction with calmodulin, one in the submicromolar and the other in the micromolar range of Ca2+ concentrations (1). In fact, calmodulin interacts with fragment (b) on the TMEM16A protein, corresponding to the selectively spliced exon 6b, and modulates channel activity (1).

Calmodulin can interact with compounds such as 1-EBIO, DCEBIO and riluzole, that induce the opening of Ca2+-activated K+ channels with low and medium conductivities and can also activate TMEM16A (15). The TMEM16 proteins link Ca2+ signals with cellular electrical activity and lipid transport and play a critical role in CF (16). The aforementioned compounds within amino acid supplement tablets can activate the efflux of Ca2+ from cells in CF (15). Moreover, calmodulin can interact with and regulate the activity of Ca2+ channels (15). Although several structural analyses of the TMEM16 proteins, including fungal and mouse orthologues, have been completed, the binding sites for lipid scramblase activity are yet to be identified. The TMEM16 proteins that function as phospholipid scramblases have an essential role in almost all human physiological processes (2,17). Therefore, dysregulated activity of TMEM16 as a phospholipid scramblase may lead to unfavorable consequences. In summary, although these structural and functional studies provide important insights into the voltage-dependent activation mechanisms of TMEM16A as a CaCC, further studies are needed to comprehensively understand the dual functionalities of TMEM16 proteins as ion channels and phospholipid scramblases.

Characteristics of the different TMEM16 subtypes

In clinical practice, prognostic markers can predict the poor clinical outcomes of treatment methods in patients with cancer. However, the ambiguity in the molecular functions of these markers makes the accurate prediction of the progression of tumors difficult. TMEM16 protein is not a prognostic marker for tumors, but it is closely related to the occurrence and development of tumors (18). TMEM16 proteins are distributed throughout the human body, with different types distributed in different tissues or organs, and are associated with various diseases (Table I). TMEM16A and B function as both CaCCs and phospholipid scramblases that promote the bidirectional mobility of membrane lipids (6). Additionally, TMEM16A and B control the release of Ca2+ stored in the cytoplasmic membrane, enhance intracellular Ca2+ signaling, amplify Ca2+ signaling activated by G protein-coupled receptors and regulate ion channel trafficking (6). TMEM16A is mainly involved in trans-epithelial Cl transport (1,4,19) and smooth muscle tone regulation (20-22), and is widely expressed throughout the body, serving as a receptor to sense injury stimuli and cell proliferation (particularly when upregulated in cancer). In addition, an induction of the production of angiotensin II stimulates the contraction of cerebral vessels via the TMEM16A-mediated Ras homolog family member A/Rho-associated protein kinase signaling pathway (23). Furthermore, the P38/JNK signaling pathway is also activated by TMEM16A expression, thereby increasing the apoptosis rate of podocytes in mice with diabetic nephropathy, which can exacerbate the injury caused to the kidneys (24). TMEM16A regulates the proliferation of the epithelial cells lining the bile duct via the ATP-stimulated-Ca2+-protein kinase C signaling pathway to induce the synthesis and secretion of bile (25). TMEM16B regulates sensory processes such as smell and vision and can control the excitability of neuronal and glial cells (26-28). TMEM16B mutations can cause multiple sclerosis and schizophrenia (4,29-31). TMEM16C is typically expressed in the central and peripheral nervous systems of humans, mice and rats, and interacts with Na+-activated K+ channels to improve the susceptibility of Na+ and the activity of K+ channels (32-34). TMEM16C has a role in certain other cellular functions including the regulation of pain and heat processing (34). Previous studies have revealed that genetic mutations in TMEM16C can cause craniocervical dystonia in humans (35-37). TMEM16D also functions as a non-selective ion channel and a phospholipid scramblase (38), is mainly expressed in the brain and endocrine glands, and it can control the mean arterial pressure and secrete aldosterone (39). A mutation in the gene encoding TMEM16D can lead to neurological diseases, such as Alzheimer's disease (40). TMEM16E acts as a non-selective ion channel and scramblase, and is mainly expressed in skeletal muscle, participating in the repair and maintenance of intracellular calcium stability of skeletal muscle, and activates the janus kinase (JAK)/STAT3 signaling pathway for cell migration and invasion (12). TMEM16E causes gnathodiaphyseal dysplasia (GDD) in cases of missense mutations (41) and muscular dystrophy (MD) in cases of functional mutations (42-44). TMEM16F also acts as a non-selective ion channel and scramblase activated by very high concentrations of Ca2+ and promotes the translocation of phospholipid and phosphatidylserine (PS) from the inner leaflet of the plasma membrane to the outer leaflet (45,46). Mutations in the gene encoding TMEM16F are associated with the development of Scott syndrome, a hemorrhagic disease caused by phospholipid-related disorders in the membranes of platelets (7,47,48). Moreover, TMEM16F mediates the proliferation of myoblasts; it plays an essential role in C2C12 myoblast proliferation, likely via regulating the ERK/AKT signaling pathway (49). The roles of TMEM16G and H have not yet been fully elucidated (50). The levels of TMEM16G are upregulated in cancer, particularly prostate cancer, and interact with the other upregulated proteins such as intracellular vesicle proteins (51). Hence, TMEM16G may be a potential biomarker for diagnosis and a target for prostate cancer immunotherapy (51). TMEM16G is also involved in the perturbation of the lipid bilayer in cell lines with a deletion in TMEM16F (7,50). TMEM16H forms junctions between the endoplasmic reticulum (ER) and the cell membrane in intracellular signaling and is involved in the transport of bile salts and the manifestation of intrahepatic cholestasis of pregnancy (52,53). The intrinsic process of TMEM16H may involve an interaction between proteins such as the matrix-interacting molecule 1, and receptors such as the inositol 1,4,5-trisphosphate receptor, that induce the release of Ca2+ from the cells (52). TMEM16J is a non-selective cation channel with scramblase activity (54,55), is activated by cAMP-dependent protein kinase A (5) and is associated with the development of certain types of cancer, such as gastric cancer, pancreatic cancer and esophageal squamous cell carcinoma (56-58). TMEM16K is mainly localized to the membranes of intracellular compartments, is the most studied phospholipid scramblase and demonstrates non-specific ion channel activity that is optimally regulated by Ca2+ and short-chain lipids (59). TMEM16K is involved in spindle assembly (60) and affects macrophage volume regulation (61). TMEM16K deficiency leads to spinocerebella ataxia autosomal receiving type 10 (59). In addition, TMEM16K also forms contact sites with endosomes and is associated with Ca2+ signaling, cell volume regulation and apoptosis (59-61).

Table I

Functions, expression, physiological role and diseases related to TMEM16 proteins.

Table I

Functions, expression, physiological role and diseases related to TMEM16 proteins.

ProteinFunctionsExpressionPhysiological roleRelated disease
TMEM16A CaCC/scramblaseWidely expressed.Transepithelial ion transport, smooth muscle contraction, activation of nociceptive neurons and cell proliferation and invasion.GIST, HNSCC, pancreatic ductal adenocarcinoma, gastric cancer, colon cancer and esophageal cancer.
TMEM16B CaCC/scramblaseSynaptic terminals of light and olfactory receptors.Regulates olfactory and visual senses and controls excitability of neurons and glial cells.Multiple sclerosis and schizophrenia.
TMEM16CScramblaseCerebral cortex, cerebellum, hippocampus and caudate body.Enhancing the K/Na+ channel and regulation of pain and heat processing.Cluster headaches, Huntington's disease, osteoarthritis, eczema and craniocervical muscle tone disorder.
TMEM16DNon-selective cation channel/scramblaseEndocrine tissue and the brain.Aldosterone secretion and cell proliferation.Severe hypoglycemia, Alzheimer's disease and multiple sclerosis.
TMEM16ENon-selective ion channel/scramblaseGastrointestinal tract and skeletal muscles.Cell migration and invasion by affecting the JAK/STAT3 pathway, maintaining cytosolic Ca2+ homeostasis and muscle repair.Mutated in GDD, LGMD, and MMD.
TMEM16FNon-selective ion channel/scramblaseEndothelial cells.Programmed cell death, lymphocyte apoptosis and activation of coagulation system.Scott syndrome and amyotrophic lateral sclerosis.
TMEM16GIon channel?/scramblaseCartilage, spleen, prostate and mainly in the gastrointestinal tractInteractions with intracellular vesicle proteins.Prostate cancer, breast cancer, familial non-nodular thyroid cancer and neuroblastoma
TMEM16HCaCC?Expression level is relatively low in most organisms.Connecting the plasma membrane and endoplasmic reticulum, intracellular signal transduction and bile salt transport.Intrahepatic cholestasis of pregnancy.
TMEM16JcAMP/PKA-activated cation channel/scramblaseThe plasma membrane of olfactory epithelium, the endoplasmic reticulum of renal epithelial cells and the brush like plate membrane of proximal tubular epithelial cells.Inducing cell proliferation, apoptosis and invasion and upregulating calcium influx.Chronic kidney disease, pancreatic cancer and colorectal cancer.
TMEM16KScramblaseBetween two endoplasmic reticulum lobules.Redistribution of phosphatidylserine, regulation of macrophage immune function, cell proliferation and apoptosis.Spinocerebellar ataxia and CoQ10 deficiency.

[i] GIST, gastrointestinal stromal tumor; HNSCC, head and neck squamous cell carcinoma; CaCC, Ca2+-activated Cl channel; PKA, protein kinase A; GDD, gnathodiaphyseal dysplasia; LGMD, limb girdle muscular dystrophy; MMD, Miyoshi muscular dystrophy; JAK, janus kinase.

Expression levels of TMEM16 proteins and their applicability as therapeutic targets in different diseases

Association between TMEM16 and CF
CF

CF is a genetic disease that affects multiple organs. It is caused by abnormal CFTR transport through the epithelial layer and is characterized by a loss of function in various systems. This disease, caused by mutations in a single gene encoding CFTR, can shorten the lifespan of humans (62,63). Patients with CF present with symptoms that indicate the effects on a wide range of organs in the body (62). These include obstruction of the ducts of the mucinous glands and changes in membrane composition in lung epithelium (62). However, certain non-malignant types of CF are virtually asymptomatic and are diagnosed only in adulthood; they affect only a single organ, as there is no systemic involvement (63). The more severe types of CF cause afflictions that affect multiple organs, including male infertility, severe respiratory dysfunction (including bronchiectasis, emphysema and pulmonary edema) and pancreatic and intestinal complications (64). The progressive deterioration of lung function and multiple organ failure are the main causes of mortality in patients with CF (65). Furthermore, multiple therapies have been clinically approved (NCT04058366) for treating the individual conditions associated with CF (66-68). However, novel modulators, multiple experimental approaches and advanced cellular models in patients with CF should be developed to accurately and reliably predict the drugs in clinical settings.

Regulatory mechanism of CFTR

CFTR, a transmembrane conductance regulator protein with 1,480 amino acids, is a Cl channel driven by cAMP (64) and is located in the apical membrane of secretory epithelial cells. CFTR can transport both Cl and HCO3 into epithelial cells. The channel is an ATP-binding transporter comprising five domains: Two transmembrane domains that form the channel pore, a regulatory domain (R) and two nucleotide-binding domains (NBD1/NBD2) (69). High levels of Cl are necessary for the important physiological actions of CFTR in the epithelial cells of the airways, including moistening the mucosal surface and removing the mucosal cilia (63,69). Patients with CF lack CFTR, which causes mucus aggregation and tracheal blockage and leads to susceptibility to chronic bacterial infections (63). These patients are therefore at risk of respiratory diseases. In addition, these airway epithelial cells also express TMEM16A, which functions as a second Cl channel; the cytosolic Ca2+ concentrations control its activity, and several inhibitors or agonists have been identified (69) (Fig. 3).

In total, ~90% of patients with CF harbor a mutation referred to as F508del, which leads to the degradation of proteases and retention of the ER. A minimal increase in the occurrence of F508del in the CFTR gene was observed in the plasma membrane of apical cells (66,70,71). In total, ~50% of patients with CF are homozygous for F508del, which not only have a processing defect but also significantly reduces the stability and flexibility of the cell surface in channel gating, if the mutation affects CFTR localization to the plasma membrane (66). In such patients, administering lumacaftor as a monotherapy may reduce the levels of Cl in the sweat by up to 8 mmol/l, in a dose-dependent manner. However, lumacaftor failed to improve abnormal lung function in phase II trials (71-73). In an in vitro preclinical trial (NCT01225211), the addition of high concentrations of ivacaftor as an enhancer was twice as effective as lumacaftor alone, achieving ~25% of the normal CFTR activity (71). Thus, adding enhancers significantly improved lung dysfunction (predicted forced expiratory volume value of 1%) by 3-4% and reduced lung functionality deterioration rates (71,73,74). In particular, the combined use of two corrective agents and one enhancer [elexacaftor-tezacaftor-ivacaftor (Trikafta) combination, NCT03525444] was significantly effective in treating the most commonly manifested defects in the membrane transport and gating caused by CFTR mutations (69,70,75). The addition of tezacaftor, a corrective agent, for treating patients homozygous for F508del markedly improved diminished respiratory function (70). Administration of Kalydeco®, a pharmacokinetic enhancer, can be used to restore the damage caused to gated membrane transport by CFTR channels due to missense mutations in CFTR (66,74,76).

TMEM16A as a therapeutic target for the treatment of CF

The known modulators of TMEM16A, including CACCinh-A01, Fact, Eact, MONNA, TMinh-23, T16inh-A01, ETX001, Ani9 and phenyl quinoxalinone (CFTRact-J027) are either inhibitors or activators (Fig. 4) (77). Phenyl quinoxalinone is used to treat conditions such as constipation, dry eyes, cholestatic liver and inflammatory lung diseases; it is a highly effective modulator of CF and can thus change its course and progression (77). This type of CFTR-modulating drugs are potential candidates for use in novel therapeutic methods of CF (77). They can enhance the expression of the mutated gene in the G551D-CFTR mutant to the original functional level and even restore it to the pre-mutation state (66). Notably, TMEM16A may demonstrate functions in addition to the secretion of electrolytes and mucus and further engage in the proliferation of basal cells and the repair mechanisms of epithelial cells (78,79).

Before the discovery of TMEM16A, evidence suggested the occurrence of a second Cl-channel expressed in the epithelial cells of the airways of patients with and without CF (80). This second channel, referred to as a CaCC, is regulated by the cellular concentrations of the Ca solute. Stimulation of the apical membranes of the epithelial cells of the airways with the purinergic agonist ATP in vitro and in vivo elicited a large but transient Cl secretory response (16,81,82). The putative physiological role of CaCCs in the epithelial cells of the airways can involve mechanical stimuli, such as those caused by normal tidal breathing or coughing, that can induce the release of ATP, thereby promoting the secretion of Cl by the mucosal layer of the airways through the binding of ATP to the purinergic and CaCC-associated receptors and finally triggering the influx of Ca2+ (21,83). The secretion of mucus in the airways involves TMEM16A (84).

By contrast, TMEM16A plays a key role in the movement of the tracheal cilia and reducing the discharge of mucus (85). TMEM16A simultaneously guides chlorine gas and bicarbonate through the airway epithelium and is expressed in the surface epithelium and submucosal glands, removing mucosal cilia by enhancing anion influx (85). In addition, the inhibition of TMEM16A by pharmacological compounds reduced the production of fluids on the surfaces of the airways (84). For instance, niclosamide reduced mucus production in the airways of sensitized mice (16) and it was reported to also affect intracellular calcium homeostasis by inhibiting the SERCA calcium pump (86). Therefore, the reported effects, such as the inhibition of mucus and cytokine release, bronchodilation and antibacterial activity, make niclosamide a potential drug suitable for the treatment of inflammatory diseases of the airways such as CF, asthma and chronic obstructive pulmonary disease (86). A recently identified TMEM16A potentiator, ETX001, triggers the secretion of fluids and accelerates the mucus clearance process without causing bronchoconstriction (77,87). The structure, the detailed mechanism of action, the location of the binding site and the selectivity profile of ETX001 have not yet been reported, although ETX001 may not interfere with Ca2+ signaling (77). Moreover, the functional efficiency of CFTR in epithelial cells can be improved by blocking microRNA (miR)-based RNA silencing and post-transcriptional regulation to increase the expression of TMEM16A (88).

Although TMEM16A may represent a potential target for the pharmacotherapy of CF, the widespread expression of TMEM16A is a serious concern since systemic administration may produce a broad range of side effects. Thus, any treatment targeting TMEM16A requires selective treatment regimens using specific drugs.

Roles of TMEM16 in diverse tumor types
Development and proliferation of tumors mediated by TMEM16 proteins

Tumor growth is critically associated with cell differentiation and proliferation. The regulation of the intracellular Ca2+ levels by the TMEM16 proteins may affect tumor development or regulate the exocytosis of the cell membrane by controlling the intracellular concentrations of Cl (3,11). The activation of CaCCs by cellular Ca2+ mainly occurs in the proliferative potential cells and different types of cancer cells (89). The expression levels of TMEM16 proteins in diverse types of cancer, including TMEM16A-mediated gastrointestinal stromal tumor (18), leiomyosarcoma (90), head and neck cancer (91), carcinoma of the lungs (92), pancreatic cancer (93), prostate cancer (94), breast cancer (95), colorectal cancer (96), gastric cancer (97), glioma and glioblastoma (98), esophageal cancer (99) and chondroblastoma (100), are indicated in Table II. TMEM16A participates in cancer proliferation and migration by influencing the MAPK and Ca2+/calmodulin-dependent protein kinase (CAMK) signaling pathways and interacts with epidermal growth factor receptor (EGFR) in head and neck squamous cell carcinoma (HNSCC) (91). TMEM16E promotes the development of colorectal (92) and thyroid (101) cancer. TMEM16G promotes the development of prostate (51) and breast (102) cancer, and TMEM16J promotes the development of pancreatic cancer (57). Therefore, ascertaining the links between TMEM16 proteins and pathways or mechanisms in tumor cells is important for inhibiting tumor growth and proliferation and developing therapeutic methods in clinical settings. However, to the best of our knowledge, all commercially available TMEM16 protein detection kits are for research purposes only and not for clinical practice. Due to a lack of availability of antibodies against the human-derived TMEM16 proteins and significant interspecific differences in the sequences of the TMEM16 proteins, cross-reactivity is unlikely to occur. Thus, the development of a TMEM16 detection kit for the diagnosis of cancer requires the production of antibodies against the TMEM16 protein in humans.

Table II

Cancer or physiological processes mediated by various TMEM16 proteins.

Table II

Cancer or physiological processes mediated by various TMEM16 proteins.

TMEM16Cancer or process(Refs.)
TMEM16Agastrointestinal stromal tumor(18)
Leiomyosarcoma(90)
Head and neck cancer(91)
Lung cancer(92)
Pancreatic cancer(93)
Prostate cancer(94)
Breast cancer(95)
Colorectal cancer(96)
Gastric cancer(97)
Glioma, glioblastoma(98)
Esophageal cancer(99)
Chondroblastoma(100)
TMEM16BRegulate olfactory sensation and vision(29)
Control the excitability of neurons and glial cells(31)
TMEM16CCraniocervical dystonia(93)
TMEM16DControl of mean arterial pressure(39)
TMEM16EColorectal cancer(92)
Thyroid cancer(101)
TMEM16FMyoblast proliferation(49)
TMEM16GProstate cancer(51)
Breast cancer(96)
TMEM16HParticipate in the formation of the connection between ER and cell membrane(52,53)
TMEM16JGastric cancer(56)
Pancreatic cancer(57)
TMEM16KSpindle formation(60)
Volume regulation(61)

In summary, TMEM16 can regulate the biochemical/molecular processes in tumors, and its abnormal expression in malignant tumors provides the possibility of employing it as a clinical biomarker for early diagnosis and a therapeutic target for reducing the occurrence and/or growth of tumors.

Various signaling pathways of TMEM16A in tumors

TMEM16A, in conjunction with EGFR, mediates the growth of tumors through two routes. First, as a CaCC, TMEM16A promotes the expression of cyclin D1 (CCND1) via the CAMK and AKT signaling pathways in succession, thereby leading to tumor proliferation (103). Second, TMEM16A participates in cancer proliferation and migration by influencing the MAPK and CAMK signaling pathways. Subsequently, the MAPK signaling pathway activates the MAPKK signaling pathway, which binds to the Grb2 binding site on EGFR with son of sevenless protein on Ras protein, altering the synthesis and activation of the Raf protein, which subsequently activates CCND1 via the phosphorylation of MEK (104). Furthermore, activation of the MAPKK signaling pathway promotes angiogenesis in vascular endothelial cells (104). As EGFR regulates the expression levels of tumor-associated genes, different signaling pathways mediate the development of several types of cancer (105-107). For instance, the development of glioma is mediated by activation of the NF-κB signaling pathway (108,109) and HNSCC is mediated by the Ras/Raf/MEK/ERK1/2 signaling pathway (110). Another set of signaling pathways, p38 and ERK1/2, are associated with promoting hepatocarcinogenesis (Fig. 5) (111).

The number of chromosomal amplicons, the extent of promoter methylation and miRs regulate the expression levels and functions of TMEM16A. The amplicon in chromosome 11q13 consists of TMEM16A-encoding and apoptosis-related genes, such as FAS-associated death domain protein (112). Upon expression, the amplicon drives the proliferation of cancer cells. Hypermethylation of the TMEM16A gene promoter promotes the metastasis of cancer cells but inhibits their proliferation. By contrast, hypomethylation enhances the proliferation of cancer cells but inhibits their metastasis (113). miR-132 (114), miR-9 (115) and miR-381 (97) directly target the mRNAs of TMEM16A, of which miR-381 downregulates epithelial-mesenchymal transition by suppressing the TGF-signaling pathway, thereby inhibiting germinal center cell proliferation and metastasis (97). In addition to the downregulation of these miRs, the levels of TMEM16A are upregulated by transcription of the genes associated with the IL4/IL13/JAK/STAT3/STAT6 axis, thereby activating histone deacetylase, enhancing the production of steroids such as testosterone, removing cells from their physiological environment, cellular reorganization and promoting mitosis (3,116,117).

Mechanism of action of the drugs targeting TMEM16 proteins

The use of niclosamide, a potent TMEM16A inhibitor that suppresses the expression of NF-κB and the Wnt/β-catenin, IL-6/JAK1/STAT3 and GSK-3 signaling pathways, has been approved for use by the US Food and Drug Administration (83,118-120). Niclosamide not only controls the cell cycle by activating the Let-7d/CDC34 axis (121), but also by blocking the Notch signaling pathway in addition to inhibiting goblet cell metaplasia in asthmatic mice (121-125). Tumorigenesis is also inhibited either via knockdown of the TMEM16A encoding gene or the exogenous administration of low concentrations of TMEM16A (126-128). Thus, the antiproliferative effects of niclosamide are associated with its inhibitory effects on TMEM16A and have been used in clinical trials in patients with prostate and colorectal cancer (86,94,119,129). In summary, the various anticancer effects of niclosamide may be related to inhibiting the multiple cancer-promoting mechanisms of TMEM16A.

Bee venom is a complex mixture of natural products such as peptides, enzymes, bioactive amines and non-peptide components with various pharmacological properties (130,131). Bee venom peptide is a potent activator of TMEM16; it promoted the Cl currents in cells overexpressing the genes encoding TMEM16A, F, J and K; these proteins can also stimulate phospholipase A2 (PLA2) (7,11,132). Furthermore, reactive oxygen species (ROS) and lipid peroxidation can activate the TMEM16 proteins (133). The enhanced production of ROS and its associated lipid peroxidation causes ferroptosis (134), which is mainly characterized by iron-dependent lipid peroxide damage-induced cell death occurring in the mitochondria. Lipid peroxidation can be induced by erastin inhibition of cysteine import through the transporter system Xc, which leads to the depletion of glutathione and the inactivation of glutathione peroxidase (135). The death of cancer cells can be induced by bee venom peptide, and PLA2-dependent activation of metalloproteinase is essential for this effect (132). Therefore, the bee venom peptide promotes the iron-dependent death of cells, including cancer cells, a process in which TMEM16A and F are activated, in turn leading to the activation of PLA2 (132,136), which then finally induces the death of cancer cells (133,137). A number of studies have unraveled the underlying mechanisms and supported the potential therapeutic applications of TMEM16 protein, but its side effects on the human body still need further study.

Association between TMEM16 proteins and cardiovascular disease

TMEM16F is the most widely expressed TMEM16 protein that functions as both an ion channel and a phospholipid scramblase and plays a significant role in several physiological processes of various cells (46). The PS that arises on the surface of the activated platelets necessitates the involvement of phospholipid scramblases, such as TMEM16F, in the formation of the thrombin and prothrombin complex (138). Hence, TMEM16F-mediated exposure to PS is an important process in platelet aggregation and release to the blood (139). TMEM16F and its closest paralog, TMEM16E, both support coagulation on endothelial cells via PS externalization (138,140). As aforementioned, mutated TMEM16F protein can cause Scott Syndrome, a hemorrhagic disease with symptoms such as defects in blood coagulation, long-term bleeding and thrombosis (141). TMEM16F is a Ca2+-activated non-selective channel, which plays an essential role in the exposure of PS and the repair of the plasma membrane after pore formation (141-143). Therefore, TMEM16F may be a target for the innovation of novel drugs that can help treat hemostasis and thrombotic diseases (such as stroke and heart attack) in humans. In addition, endothelial cells play a thrombotic role in hyperuricemia via the TMEM16F-mediated exposure of PS and the release of particulate molecules into the blood (144).

Association between coronavirus disease 2019 (COVID-19) and TMEM16 proteins

Formation of syncytia in COVID-19

As the name suggests, COVID-19 was first identified in 2019 and has become a serious worldwide health concern due to the large number of fatalities. To reduce the mortality rate in critical patients of COVID-19, targeted therapeutics are continuously being developed based on biological and etiological characteristics. COVID-19 causes severe respiratory conditions, such as pulmonary edema and thrombosis, acute respiratory distress syndrome and other diseases (145-147). The development of syncytia in the lungs of patients is a characteristic feature of infection by coronavirus. The host cell acts on angiotensin-converting enzyme 2 (ACE2) to activate the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein in a two-step hydrolytic process (148). The first step involves breaking the spike between the S1 and S2 subunits of the spike protein before or after binding to the receptor. The second step involves the hydrolysis and exposure of the S2 subunit, which immediately binds to the cell membrane and generates a protease to invade normal lung cells (149). Infected lung cells present with a multinucleated and abnormal morphology; SARS-CoV-2 and Middle East Respiratory Syndrome Coronavirus can fuse with the cells expressing the relevant receptors to form syncytia (150).

Mechanisms underlying the interaction of the SARS-CoV-2 spike protein with TMEM16 proteins

The cells that form syncytia and express the SARS-CoV-2 spike protein on their surface demonstrate increased concentrations and enhanced oscillations in the levels of Ca2+ along with high expression of the Ca2+-activated TMEM16 proteins in the cytoplasmic membrane, resulting in the relocation of PS and secretion of Cl (149). The associations between the levels of TMEM16, the levels of Ca2+ and the activation of TMEM16 by the SARS-CoV-2 spike protein may enhance the magnitude of Ca2+-based signaling spontaneously (150). At least three mechanisms explaining the activation of TMEM16 have been proposed. The first involves the direct cis-binding and activation of spike-protein-expressing cells, the second involves trans-binding and the initiation of protease activity via binding to ACE2 and the third involves indirect activation by inducing the release of Ca2+ (150). TMEM16F can significantly reduce calcium oscillations and membrane conductivity in spike expressing cells, used to expose PS on the cell surface (150). However, the overexpression of TMEM16F significantly stimulated the SARS-CoV-2 spike protein-induced formation of syncytia. Therefore, the expression of the spike protein, which is required for the formation of syncytia, can be reduced by the downregulation of TMEM16F. Thus, TMEM16F is identified as the major bifunctional cell membrane-phospholipid scramblase and Ca2+ channel these cells (Fig. 6).

TMEM16 participates in COVID-19 related coagulopathy

In patients affected by COVID-19, inflammation-induced damage to endothelial cells may lead to the release of a large amount of plasmin activator, thus producing high concentrations of D-dimers and degradation products of fibrin. A specific cytokine storm composed of high concentrations of proinflammatory cytokines and chemokines occurs in the body (146). These proinflammatory factors include TNF-α, IL-1 and IL-6. TNF-α and IL-1 are the primary mediators driving the inhibition of the endogenous anticoagulant pathway (147). IL-6 can induce the expression of tissue factors on monocytes, subsequently initiating the activation of coagulation and generation of thrombin (150). The PS on the surface of the activated platelets requires the activity of the phospholipid scramblase to participate in the formation of the thrombin and prothrombin complex, and the TMEM16F-mediated exposure of PS is crucial for the aggregation of platelets and release to the blood (151). The activation of TMEM16F increases with the formation of syncytia, enhances the aggregation of platelets and utilization of the coagulation factors, which leads to the production of an abnormal amount of thrombin and fibrin and finally causes disseminated intravascular coagulation (DIC) and microangiopathy (146-151). The involvement of TMEM16F in COVID-19-related disorders in blood coagulation results in a combination of low-grade DIC and local, pulmonary and thrombotic microvascular disease, which may significantly impact the dysfunction of the organs in the most severely affected patients.

Mechanisms underlying a novel coronavirus treatment method using drugs targeting TMEM16

Specific drugs should be developed to combat COVID-19 and to provide strategies for treating similar coronaviruses by analyzing the mechanisms behind the formation of viral syncytia and mining for the drugs acting on the Cl channels. Niclosamide, a drug that suppresses the formation of syncytium by inhibiting TMEM16F, has been identified as a promising drug for the treatment of severe COVID-19 and has been approved for use by the US Food and Drug Administration (150,152). Niclosamide is highly hydrophobic and therefore has poor solubility in aqueous solution (153). The hydrophobic helix formed by the TM1-6 grooves in both TMEM16A and F and the residues in this helix are essential for drug binding (11). TM6 functions as the main gating element of the channel and is part of the ion-conduction pore formed by TMEM16A and F (2,154). The antagonist simultaneously locks both of these ion-conduction pores by binding to the upper region of the TM6 in a closed configuration. In addition to niclosamide, nitazoxanide and 1PBC bind to the same conserved sites (11,155).

Furthermore, hexachlorophene, dichlorophenol, gefitinib (156), trifluoperazine (16), serotonin reuptake inhibitors and ivermectin (157), which target TMEM16A, can also inhibit the spike protein-induced formation of syncytia. Thus, screening specific drugs may reveal a common mechanism underlying the spike protein-dependent cell-cell fusion. Since the possibility of the recurrence of another COVID-19 pandemic is high, drug development is an indispensable part of future research.

Role of TMEM16 proteins in orthopedics

TMEM16A as a marker of osteoporosis

Osteoporosis is a systemic disease resulting in a decrease in the mineral density and quality of the bones due to various causes, thus leading to changes in the microstructure and increased fragility of bones, thereby leading to fractures (158). TMEM16A is directly regulated by the cytosolic concentrations of Ca2+ and indirectly by interactions with calmodulin (12). Osteoporosis causes changes in the Ca2+ concentrations, which affects the levels and function of TMEM16A and therefore can be used as a marker for the early diagnosis and targeted intervention of osteoporosis. Furthermore, a deletion in the gene encoding TMEM16A caused severe defects in the tracheal cartilage in mice, which could be fatal in certain cases (159). In addition, the TMEM16A blockers, benzbromarone and CaCCinh-A0, 1 can significantly inhibit the differentiation of osteoclasts (159).

TMEM16E expression levels in bones and muscles

TMEM16E is a 913 amino acid protein with a Ca2+-activated phospholipid scramblase activity (160). TMEM16E is significantly expressed in muscles and bones, with Ca2+ activation involving TM4 and 5 (41). Mutations in the gene encoding TMEM16E in humans are associated with two genetic disorders: GDD, a rare skeletal-system-related syndrome associated with bone deformity and increased fragility (161), and limb-girdle MD (LGMD), a type of progressive MD (162,163). A recent model demonstrated that GDD is characterized by the manifestation of tubular bone sclerosis and mandible bone lesions, including those of a giant jawbone, arched tibia, fragility, sclerosis and cortical thickening of the femoral and tibial epiphysis (162). In the blood culture of this model, the number of osteoblasts increased while the number of osteoclasts decreased (162). The p.Cys360Tyr mutation in TMEM16E inhibits receptor activator of NF-κB ligand-induced p65, Erk, p38 and AKT phosphorylation, implying that multiple signal transduction processes contribute to osteoclast maturation and bone resorption (164). A recent study has assisted in generalizing the correlations between the GDD-associated genotype and the phenotype by extending the observations made to the GDD mutants of TMEM16E such as p.Arg215Gly, p.Cys356Gly/Arg/Tyr, p.Cys360Tyr, p.Ser500Phe and p.Gly518Glu (42). In the proposed model, p.Cys360Tyr causes osteosclerosis with a high bone turnover and can be a potential target for treating GDD (Fig. 7).

Mutations in the TMEM16 gene can lead to different orthopedic genetic-related diseases

TMEM16E is highly associated with TMEM16F; both are dual-function proteins with non-selective ion channel and phospholipid scramblase activities. In addition to participating in the exposure of PS and promoting different physiological processes, TMEM16E is also involved in bone mineralization and skeletal muscle repair (11). Expression of TMEM16E is highest in the cardiac and skeletal muscles and in growth plate chondrocytes and osteoblasts (43). Dysfunctional TMEM16E protein causes bone dysplasia and fragility and suppurative osteomyelitis of the lower jaw; in addition, LGMD-related amino acid substitutions cause a loss of function, while GDD-related substitutions lead to a constitutive lipid scramblase activity without the requirement of elevated cytosolic Ca2+ levels (42). The dysregulation of TMEM16E also causes arthritis and MD (162,163). Mutated TMEM16C protein causes muscle-related diseases associated with involuntary muscle spasms caused by cranial-neck dystonia (165). The discovery of the relationship between GDD-related mutations and the functional phenotypes confirmed the speculation that interventions based on genetic patterns may greatly improve the treatment outcomes of genetic diseases in humans, which is of great significance for the health and survival of affected individuals. Therefore, it is crucial to understand the mechanism of action of TMEM16 in genetic-related orthopedic diseases and to decipher the molecular mechanisms underlying the activation of gating and regulatory functions to develop novel targeted drug-based therapies towards the early intervention of diseases in the future.

Conclusions and future perspectives

In summary, the TMEM16 proteins have emerged as important pharmacological targets for the treatment of several associated diseases. Since the discovery and identification of TMEM16 as a Cl channel, its roles in various human diseases have been established. The availability of a large amount of structural information has led to significant progress in understanding the role of TMEM16 in disease progression at the molecular level. Thus, its role in the pathogenesis of human diseases, research involving the therapy of such diseases based on the targeting of TMEM16 and understanding the mechanisms of action of the proposed drugs with an enhanced potential should be addressed further. The studies on the structural characteristics of TMEM16 have led to the revelation of its various functions, ranging from ion transport to the modulation of the dynamics of the plasma membrane phospholipids and the underlying molecular mechanisms. However, further research is needed to fully understand the roles played by the other anoctamins besides TMEM16A and B. This requires efforts based on multiple methods, including gene silencing in wild-type cells, overexpression in hematopoiesis systems and generating conditional gene knockouts in mice.

Based on the various research models, such as the 'modular design' model explaining TMEM16 assembly and the 'clam-shell' and the 'pore-dilation' gating/permeation models elaborating the scramblase and channel activities of TMEM16, it is possible to address: i) The study and analyses of the synergistic effects of the three Ca2+-binding sites on the TMEM16 protein under normal physiological environments; ii) a more in-depth analysis of the mutated sites in the gene encoding the TMEM16 protein that is related to the manifestation of genetic diseases in humans to develop novel therapeutic drugs with improved specificity and targeting; and iii) the requirement of the careful designing of such drugs to avoid side effects as TMEM16A, F and other anoctamins are expressed in multiple tissues of the human body. It is thus hoped that future research on the structural properties of TMEM16 proteins can capture the open conformations and assist the designing of potential drugs that target Cl channels via utilizing the existing models more comprehensively and promoting development and innovation in this field.

Availability of data and materials

Not applicable.

Authors' contributions

ZH, ZI, ZZ, XC, AM, JL, WL and ZD participated in the literature review, ZH and ZD performed the figure design and ZH wrote the manuscript. ZH, ZI and ZD revised the paper. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent for participation

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

Wencui Li ORCID ID: 0000-0003-2787-5360. Zhiqin Deng ORCID ID: 0000-0002-0819-8504.

Abbreviations:

CFTR

CF transmembrane conductance regulator

GDD

gnathodiaphyseal dysplasia

CF

cystic fibrosis

CaCCs

Ca2+-activated Cl channels

EGFR

epidermal growth factor receptor

CCND1

cyclin D1

PS

phosphatidylserine

Acknowledgements

Not applicable.

Funding

This study was supported by The National Natural Science Foundation of China (grant no. 81972085, 82172465), China University Industry-University-Research Innovation Fund (grant no. 2021JH037), The Natural Science Foundation of Guangdong Province (grant nos. 2021A1515010706 and 2023A1515010102), Guangdong Provincial Key Clinical Discipline-Orthopedics (grant no. 2000005), The Sanming Project of Shenzhen Health and Family Planning Commission (grant no. SZSM202311008), Shenzhen Science and Technology Planning (grant no. GJHZ20210705142007023) and The Shenzhen Key Medical Discipline Construction Fund (grant no. SZXK025).

References

1 

Vocke K, Dauner K, Hahn A, Ulbrich A, Broecker J, Keller S, Frings S and Möhrlen F: Calmodulin-dependent activation and inactivation of anoctamin calcium-gated chloride channels. J Gen Physiol. 142:381–404. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Whitlock JM and Hartzell HC: Anoctamins/TMEM16 proteins: Chloride channels flirting with lipids and extracellular vesicles. Annu Rev Physiol. 79:119–143. 2017. View Article : Google Scholar :

3 

Kunzelmann K, Ousingsawat J, Benedetto R, Cabrita I and Schreiber R: Contribution of Anoctamins to cell survival and cell death. Cancers. 11:3822019. View Article : Google Scholar : PubMed/NCBI

4 

Scudieri P, Sondo E, Ferrera L and Galietta LJV: The anoctamin family: TMEM16A and TMEM16B as calcium-activated chloride channels. Exp Physiol. 97:177–183. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Kim H, Kim H, Lee J, Lee B, Kim HR, Jung J, Lee MO and Oh U: Anoctamin 9/TMEM16J is a cation channel activated by cAMP/PKA signal. Cell Calcium. 71:75–85. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Khelashvili G, Falzone ME, Cheng X, Lee B-C, Accardi A and Weinstein H: Dynamic modulation of the lipid translocation groove generates a conductive ion channel in Ca2+-bound nhTMEM16. Nat Commun. 10:49722019. View Article : Google Scholar :

7 

Agostinelli E and Tammaro P: Polymodal control of TMEM16x channels and Scramblases. Int J Mol Sci. 23:15802022. View Article : Google Scholar : PubMed/NCBI

8 

Baethge C, Goldbeck-Wood S and Mertens S: SANRA-a scale for the quality assessment of narrative review articles. Res Integr Peer Rev. 4:52019. View Article : Google Scholar : PubMed/NCBI

9 

Falzone ME, Malvezzi M, Lee BC and Accardi A: Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol. 150:933–947. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Falzone ME, Rheinberger J, Lee BC, Peyear T, Sasset L, Raczkowski AM, Eng ET, Di Lorenzo A, Andersen OS, Nimigean CM and Accardi A: Structural basis of Ca2+-dependent activation and lipid transport by a TMEM16 scramblase. ELife. 8:e432292019. View Article : Google Scholar :

11 

Cheng Y, Feng S, Puchades C, Ko J, Figueroa E, Chen Y, Wu H, Gu S, Han T, Li J, et al: Identification of a conserved drug binding pocket in TMEM16 proteins. Res Sq. View Article : Google Scholar

12 

Pedemonte N and Galietta LJV: Structure and function of TMEM16 proteins (Anoctamins). Physiol Rev. 94:419–459. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Yu K, Duran C, Qu Z, Cui YY and Hartzell HC: Explaining calcium-dependent gating of Anoctamin-1 chloride channels requires a revised topology. Circ Res. 110:990–999. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Jung J, Nam JH, Park HW, Oh U, Yoon JH and Lee MG: Dynamic modulation of ANO1/TMEM16A HCO3 permeability by Ca2+/calmodulin. Proc Natl Acad Sci USA. 110:360–365. 2012. View Article : Google Scholar

15 

Tian Y, Kongsuphol P, Hug M, Ousingsawat J, Witzgall R, Schreiber R and Kunzelmann K: Calmodulin-dependent activation of the epithelial calcium-dependent chloride channel TMEM16A. FASEB J. 25:1058–1068. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Hahn A, Salomon JJ, Leitz D, Feigenbutz D, Korsch L, Lisewski I, Schrimpf K, Millar-Büchner P, Mall MA, Frings S and Möhrlen F: Expression and function of Anoctamin 1/TMEM16A calcium-activated chloride channels in airways of in vivo mouse models for cystic fibrosis research. Pflugers Arch. 470:1335–1348. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Falzone ME, Feng Z, Alvarenga OE, Pan Y, Lee B, Cheng X, Fortea E, Scheuring S and Accardi A: TMEM16 scramblases thin the membrane to enable lipid scrambling. Nat Commun. 13:26042022. View Article : Google Scholar : PubMed/NCBI

18 

Jansen K and Steurer S: DOG1 expression is in common human tumors: A tissue microarray study on more than 15,000 tissue samples. Am J Clin Pathol. 156(Suppl): S108–S109. 2021. View Article : Google Scholar

19 

Lam AK and Dutzler R: Calcium-dependent electrostatic control of anion access to the pore of the calcium-activated chloride channel TMEM16A. ELife. 7:e391222018. View Article : Google Scholar : PubMed/NCBI

20 

Huang WC, Xiao S, Huang F, Harfe BD, Jan YN and Jan L: Calcium-Activated chloride channels (CaCCs) regulate action potential and synaptic response in hippocampal neurons. Neuron. 74:179–192. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Davis AJ, Forrest AS, Jepps TA, Valencik ML, Wiwchar M, Singer CA, Sones WR, Greenwood IA and Leblanc N: Expression profile and protein translation of TMEM16A in murine smooth muscle. Am J Physiol Cell Physiol. 299:C948–C959. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Thomas-Gatewood C, Neeb ZP, Bulley S, Adebiyi A, Bannister JP, Leo MD and Jaggar JH: TMEM16A channels generate Ca2+-activated Cl-currents in cerebral artery smooth muscle cells. Am J Physiol Circ Physiol. 301:H1819–H1827. 2011. View Article : Google Scholar

23 

Li RS, Wang Y, Chen HS, Jiang FY, Tu Q, Li WJ and Yin RX: TMEM16A contributes to angiotensin II-induced cerebral vasoconstriction via the RhoA/ROCK signaling pathway. Mol Med Report. 13:3691–3699. 2016. View Article : Google Scholar

24 

Lian H, Cheng Y and Wu X: TMEM16A exacerbates renal injury by activating P38/JNK signaling pathway to promote podocyte apoptosis in diabetic nephropathy mice. Biochem Biophys Res Commun. 487:201–208. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Dutta AK, Khimji AK, Liu S, Karamysheva Z, Fujita A, Kresge C, Rockey DC and Feranchak AP: PKCα regulates TMEM16A-mediated Cl-secretion in human biliary cells. Am J Physiol Liver Physiol. 310:G34–G42. 2016.

26 

Arreola J, López-Romero AE, Pérez-Cornejo P and Rodríguez-Menchaca AA: Phosphatidylinositol 4,5-bisphosphate and cholesterol regulators of the calcium-activated chloride channels TMEM16A and TMEM16B. Adv Exp Med Biol. 1422:279–304. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Lee D, Lim H, Lee J, Ha GE, No KT and Cheong E: Intracellular loop in the brain isoforms of anoctamin 2 channels regulates calcium-dependent activation. Exp Neurobiol. 32:133–146. 2023. View Article : Google Scholar : PubMed/NCBI

28 

Pietra G, Dibattista M, Menini A, Reisert J and Boccaccio A: The Ca2+-activated Cl-channel TMEM16B regulates action potential firing and axonal targeting in olfactory sensory neurons. J Gen Physiol. 148:293–311. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Ayoglu B, Mitsios N, Kockum I, Khademi M, Zandian A, Sjöberg R, Forsström B, Bredenberg J, Lima Bomfim I, Holmgren E, et al: Anoctamin 2 identified as an autoimmune target in multiple sclerosis. Proc Natl Acad Sci USA. 113:2188–2193. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Ha GE, Lee J, Kwak H, Song K, Kwon J, Jung SY, Hong J, Chang GE, Hwang EM, Shin HS, et al: The Ca2+-activated chloride channel anoctamin-2 mediates spike-frequency adaptation and regulates sensory transmission in thalamocortical neurons. Nat Commun. 7:137912016. View Article : Google Scholar

31 

Zhang Y, Zhang Z, Xiao S, Tien J, Le S, Le T, Jan LY and Yang H: Inferior Olivary TMEM16B mediates cerebellar motor learning. Neuron. 95:1103–1111.e4. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Kim H, Kim E and Lee BC: Investigation of phosphatidylserine-transporting activity of human TMEM16C isoforms. Membranes (Basel). 12:10052022. View Article : Google Scholar : PubMed/NCBI

33 

Wang TA, Chen C, Huang F, Feng S, Tien J, Braz JM, Basbaum AI, Jan YN and Jan LY: TMEM16C is involved in thermoregulation and protects rodent pups from febrile seizures. Proc Natl Acad Sci USA. 118:e20233421182021. View Article : Google Scholar : PubMed/NCBI

34 

Huang F, Wang X, Ostertag EM, Nuwal T, Huang B, Jan YN, Basbaum AI and Jan LY: TMEM16C facilitates Na(+)-activated K+ currents in rat sensory neurons and regulates pain processing. Nat Neurosci. 16:1284–1290. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Carvalho V, Martins J, Correia F, Costa M, Massano J and Temudo T: Another twist in the tale: Intrafamilial phenotypic heterogeneity in ANO3-related dystonia. Mov Disord Clin Pract. 8:758–762. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Stamelou M, Charlesworth G, Cordivari C, Schneider SA, Kägi G, Sheerin UM, Rubio-Agusti I, Batla A, Houlden H, Wood NW and Bhatia KP: The phenotypic spectrum of DYT24 due to ANO3 mutations. Mov Disord. 29:928–934. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Esposito M, Trinchillo A, Piceci-Sparascio F, D'Asdia MC, Consoli F and De Luca A: A novel ANO3 variant in two siblings with different phenotypes. Parkinsonism Relat Disord. 111:1054132023. View Article : Google Scholar : PubMed/NCBI

38 

Reichhart N, Milenkovic VM, Wetzel CH and Strauß O: Prediction of functional consequences of missense mutations in ANO4 Gene. Int J Mol Sci. 22:27322021. View Article : Google Scholar : PubMed/NCBI

39 

Maniero C, Scudieri P, Haris Shaikh L, Zhao W, Gurnell M, Galietta LJV and Brown MJ: ANO4 (Anoctamin 4) Is a novel marker of zona glomerulosa that regulates stimulated aldosterone secretion. Hypertension. 74:1152–1159. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Sherva R, Tripodis Y, Bennett DA, Chibnik LB, Crane PK, de Jager PL, Farrer LA, Saykin AJ, Shulman JM, Naj A, et al: Genome-wide association study of the rate of cognitive decline in Alzheimer's disease. Alzheimers Dement. 10:45–52. 2014. View Article : Google Scholar

41 

Di Zanni E, Gradogna A, Scholz-Starke J and Boccaccio A: Gain of function of TMEM16E/ANO5 scrambling activity caused by a mutation associated with gnathodiaphyseal dysplasia. Cell Mol Life Sci. 75:1657–1670. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Di Zanni E, Gradogna A, Picco C, Scholz-Starke J and Boccaccio A: TMEM16E/ANO5 mutations related to bone dysplasia or muscular dystrophy cause opposite effects on lipid scrambling. Hum Mutat. 41:1157–1170. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Whitlock JM, Yu K, Cui YY and Hartzell HC: Anoctamin 5/TMEM16E facilitates muscle precursor cell fusion. J Gen Physiol. 150:1498–1509. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Foltz SJ, Cui YY, Choo HJ and Hartzell HC: ANO5 ensures trafficking of annexins in wounded myofibers. J Cell Biol. 220:e2020070592021. View Article : Google Scholar : PubMed/NCBI

45 

van Kruchten R, Mattheij NJ, Saunders C, Feijge MA, Swieringa F, Wolfs JL, Collins PW, Heemskerk JW and Bevers EM: Both TMEM16F-dependent and TMEM16F-independent pathways contribute to phosphatidylserine exposure in platelet apoptosis and platelet activation. Blood. 121:1850–1857. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Arndt M, Alvadia C, Straub MS, Clerico Mosina V, Paulino C and Dutzler R: Structural basis for the activation of the lipid scramblase TMEM16F. Nat Commun. 13:66922022. View Article : Google Scholar : PubMed/NCBI

47 

Fujii T, Sakata A, Nishimura S, Eto K and Nagata S: TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets. Proc Natl Acad Sci USA. 112:12800–12805. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Millington-Burgess SL and Harper MT: Gene of the issue: ANO6 and Scott syndrome. Platelets. 31:964–967. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Li H, Xu L, Gao Y, Zuo Y, Yang Z, Zhao L, Chen Z, Guo S and Han R: BVES is a novel interactor of ANO5 and regulates myoblast differentiation. Cell Biosci. 11:2222021. View Article : Google Scholar : PubMed/NCBI

50 

Guo J, Wang D, Dong Y, Gao X, Tong H, Liu W, Zhang L and Sun M: ANO7: Insights into topology, function, and potential applications as a biomarker and immunotherapy target. Tissue Cell. 72:1015462021. View Article : Google Scholar : PubMed/NCBI

51 

Kaikkonen E, Rantapero T, Zhang Q, Taimen P, Laitinen V, Kallajoki M, Jambulingam D, Ettala O, Knaapila J, Boström PJ, et al: ANO7 is associated with aggressive prostate cancer. Int J Cancer. 143:2479–2487. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Jha A, Chung WY, Vachel L, Maleth J, Lake S, Zhang G, Ahuja M and Muallem S: Anoctamin 8 tethers endoplasmic reticulum and plasma membrane for assembly of Ca2+ signaling complexes at the ER/PM compartment. EMBO J. 38:e1014522019. View Article : Google Scholar

53 

Liu X, Lai H, Zeng X, Xin S, Nie L, Liang Z, Wu M, Chen Y, Zheng J and Zou Y: Whole-exome sequencing reveals ANO8 as a genetic risk factor for intrahepatic cholestasis of pregnancy. BMC Pregnancy Childbirth. 20:5442020. View Article : Google Scholar : PubMed/NCBI

54 

Schreiber R, Ousingsawat J and Kunzelmann K: Targeting of intracellular TMEM16 proteins to the plasma membrane and activation by purinergic signaling. Int J Mol Sci. 21:40652020. View Article : Google Scholar : PubMed/NCBI

55 

Katsurahara K, Shiozaki A, Kosuga T, Kudou M, Shoda K, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, et al: ANO9 regulated cell cycle in human esophageal squamous cell carcinoma. Ann Surg Oncol. 27:3218–3230. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Katsurahara K, Shiozaki A, Kosuga T, Shimizu H, Kudou M, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, et al: ANO9 regulates PD-L2 expression and binding ability to PD-1 in gastric cancer. Cancer Sci. 112:1026–1037. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Jun I, Park HS, Piao H, Han JW, An MJ, Yun BG, Zhang X, Cha YH, Shin YK, Yook JI, et al: ANO9/TMEM16J promotes tumourigenesis via EGFR and is a novel therapeutic target for pancreatic cancer. Br J Cancer. 117:1798–1809. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Schreiber R, Talbi K, Ousingsawat J and Kunzelmann K: A TMEM16J variant leads to dysregulated cytosolic calcium which may lead to renal disease. FASEB J. 37:e226832023. View Article : Google Scholar

59 

Chrysanthou A, Ververis A and Christodoulou K: ANO10 function in health and disease. Cerebellum. 22:447–467. 2022. View Article : Google Scholar : PubMed/NCBI

60 

Wanitchakool P, Ousingsawat J, Sirianant L, Cabrita I, Faria D, Schreiber R and Kunzelmann K: Cellular defects by deletion of ANO10 are due to deregulated local calcium signaling. Cell Signal. 30:41–49. 2017. View Article : Google Scholar

61 

Hammer C, Wanitchakool P, Sirianant L, Papiol S, Monnheimer M, Faria D, Ousingsawat J, Schramek N, Schmitt C, Margos G, et al: A coding variant of ANO10, affecting volume regulation of macrophages, is associated with borrelia seropositivity. Mol Med. 21:26–37. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Gentzsch M and Mall MA: Ion channel modulators in cystic fibrosis. Chest. 154:383–393. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Shteinberg M, Haq IJ, Polineni D and Davies JC: Cystic fibrosis. Lancet. 397:2195–2211. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Lopes-Pacheco M, Pedemonte N and Veit G: Discovery of CFTR modulators for the treatment of cystic fibrosis. Expert Opin Drug Discov. 16:897–913. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Villamizar O, Waters SA, Scott T, Grepo N, Jaffe A and Morris KV: Mesenchymal Stem Cell exosome delivered Zinc Finger Protein activation of cystic fibrosis transmembrane conductance regulator. J Extracell Vesicles. 10:e120532021. View Article : Google Scholar : PubMed/NCBI

66 

Simon MA and Csanady L: Understanding impact of δF508 and G551D CFTR mutations on CFTR/PKA-c interaction. Biophys J. 122:112a2023. View Article : Google Scholar

67 

Harrison MJ, Murphy DM and Plant BJ: Ivacaftor in a G551D homozygote with cystic fibrosis. N Engl J Med. 369:1280–1282. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Dřevínek P, Griese M, McKone EF, Wainwright CE, Konstan MW, et al: A CFTR potentiator in patients with cystic fibrosis and theG551Dmutation. N Engl J Med. 365:1663–1672. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Fiedorczuk K and Chen J: Mechanism of CFTR correction by type I folding correctors. Cell. 185:158–168.e11. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Veit G, Roldan A, Hancock MA, Da Fonte DF, Xu H, Hussein M, Frenkiel S, Matouk E, Velkov T and Lukacs GL: Allosteric folding correction of F508del and rare CFTR mutants by Elexacaftor-Tezacaftor-Ivacaftor (Trikafta) combination. JCI Insight. 5:e1399832020. View Article : Google Scholar : PubMed/NCBI

71 

Rowe SM, McColley SA, Rietschel E, Li X, Bell SC, Konstan MW, Marigowda G, Waltz D and Boyle MP; VX09-809-102 Study Group: Lumacaftor/Ivacaftor treatment of patients with cystic fibrosis heterozygous for F508del-CFTR. Ann Am Thorac Soc. 14:213–219. 2017. View Article : Google Scholar :

72 

Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, Colombo C, Davies JC, De Boeck K, Flume PA, et al: Lumacaftor-Ivacaftor in patients with cystic fibrosis homozygous for Phe508delCFTR. N Engl J Med. 373:220–231. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Clancy JP, Rowe SM, Accurso FJ, Aitken ML, Amin RS, Ashlock MA, Ballmann M, Boyle MP, Bronsveld I, Campbell PW, et al: Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for theF508del-CFTRmutation. Thorax. 67:12–18. 2011. View Article : Google Scholar

74 

Flume PA, Harris RS, Paz-Diaz H, Ahluwalia N, Higgins M, Campbell D, Berhane I, Shih JL and Sawicki G: Long-term tezacaftor/ivacaftor safety and efficacy in people with cystic fibrosis and an F508del-CFTR mutation: 96-week, open-label extension of the EXTEND trial. J Cyst Fibros. 22:464–470. 2023. View Article : Google Scholar

75 

Bruscia EM: The effects of Elexacaftor/Tezacaftor/Ivacaftor beyond the epithelium: Spurring macrophages to fight infections. Eur Respir J. 61:23002162023. View Article : Google Scholar

76 

Sawicki GS, Van Brunt K, Booth J, Bailey E, Millar SJ, Konstan MW and Flume PA: Disease burden in people with cystic fibrosis heterozygous for F508del and a minimal function mutation. J Cyst Fibros. 21:96–103. 2022. View Article : Google Scholar :

77 

Galietta LJV: TMEM16A (ANO1) as a therapeutic target in cystic fibrosis. Curr Opin Pharmacol. 64:1022062022. View Article : Google Scholar : PubMed/NCBI

78 

Simões FB, Quaresma MC, Clarke LA, Silva IA, Pankonien I, Railean V, Kmit A and Amaral MD: TMEM16A chloride channel does not drive mucus production. Life Sci Alliance. 2:e2019004622019. View Article : Google Scholar : PubMed/NCBI

79 

Ruffin M, Voland M, Marie S, Bonora M, Blanchard E, Blouquit-Laye S, Naline E, Puyo P, Le Rouzic P, Guillot L, et al: Anoctamin 1 dysregulation alters bronchial epithelial repair in cystic fibrosis. Biochim Biophys Acta. 1832:2340–2351. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Kirk KL and Wang W: A unified view of cystic fibrosis transmembrane conductance regulator (CFTR) gating: Combining the allosterism of a ligand-gated channel with the enzymatic activity of an ATP-binding cassette (ABC) transporter. J Biol Chem. 286:12813–12819. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Deng Z, Chen X, Lin Z, Alahdal M, Wang D, Liu J and Li W: The homeostasis of cartilage matrix remodeling and the regulation of volume-sensitive ion channel. Aging Dis. 13:787–800. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Talbi K, Ousingsawat J, Centeio R, Schreiber R and Kunzelmann K: Calmodulin-dependent regulation of overexpressed but not endogenous TMEM16A expressed in airway epithelial cells. Membranes (Basel). 11:7232021. View Article : Google Scholar : PubMed/NCBI

83 

Cabrita I, Benedetto R, Schreiber R and Kunzelmann K: Niclosamide repurposed for the treatment of inflammatory airway disease. JCI Insight. 4:e1284142019. View Article : Google Scholar : PubMed/NCBI

84 

Danahay H, Fox R, Lilley S, Charlton H, Adley K, Christie L, Ansari E, Ehre C, Flen A, Tuvim MJ, et al: Potentiating TMEM16A does not stimulate airway mucus secretion or bronchial and pulmonary arterial smooth muscle contraction. FASEB Bioadv. 2:464–477. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Danahay HL, Lilley S, Fox R, Charlton H, Sabater J, Button B, McCarthy C, Collingwood SP and Gosling M: TMEM16A potentiation: A novel therapeutic approach for the treatment of cystic fibrosis. Am J Respir Crit Care Med. 201:946–954. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Ousingsawat J, Centeio R, Cabrita I, Talbi K, Zimmer O, Graf M, Göpferich A, Schreiber R and Kunzelmann K: Airway delivery of hydrogel-encapsulated niclosamide for the treatment of inflammatory airway disease. Int J Mol Sci. 23:10852022. View Article : Google Scholar : PubMed/NCBI

87 

Centeio R, Ousingsawat J, Cabrita I, Schreiber R, Talbi K, Benedetto R, Doušová T, Verbeken EK, De Boeck K, Cohen I and Kunzelmann K: Mucus release and airway constriction by TMEM16A may worsen pathology in inflammatory lung disease. Int J Mol Sci. 22:78522021. View Article : Google Scholar : PubMed/NCBI

88 

Sonneville F, Ruffin M, Coraux C, Rousselet N, Le Rouzic P, Blouquit-Laye S, Corvol H and Tabary O: MicroRNA-9 downregulates the ANO1 chloride channel and contributes to cystic fibrosis lung pathology. Nat Commun. 8:7102017. View Article : Google Scholar : PubMed/NCBI

89 

Kamaleddin MA: Molecular, biophysical, and pharmacological properties of calcium-activated chloride channels. J Cell Physiol. 233:787–798. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Sah SP and McCluggage WG: DOG1 immunoreactivity in uterine leiomyosarcomas. J Clin Pathol. 66:40–43. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Filippou A, Pehkonen H, Karhemo PR, Väänänen J, Nieminen AI, Klefström J, Grénman R, Mäkitie AA, Joensuu H and Monni O: ANO1 expression orchestrates p27Kip1/MCL1-Mediated signaling in head and neck squamous cell carcinoma. Cancers (Basel). 13:11702021. View Article : Google Scholar : PubMed/NCBI

92 

Ishaque N, Abba ML, Hauser C, Patil N, Paramasivam N, Huebschmann D, Leupold JH, Balasubramanian GP, Kleinheinz K, Toprak UH, et al: Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nat Commun. 9:47822018. View Article : Google Scholar : PubMed/NCBI

93 

Sauter DRP, Novak I, Pedersen SF, Larsen EH and Hoffmann EK: ANO1 (TMEM16A) in pancreatic ductal adenocarcinoma (PDAC). Pflugers Arch. 467:1495–1508. 2015. View Article : Google Scholar :

94 

Song Y, Gao J, Guan L, Chen X, Gao J and Wang K: Inhibition of ANO1/TMEM16A induces apoptosis in human prostate carcinoma cells by activating TNF-α signaling. Cell Death Dis. 9:7032018. View Article : Google Scholar

95 

Britschgi A, Bill A, Brinkhaus H, Rothwell C, Clay I, Duss S, Rebhan M, Raman P, Guy CT, Wetzel K, et al: Calcium-activated chloride channel ANO1 promotes breast cancer progression by activating EGFR and CAMK signaling. Proc Natl Acad Sci USA. 110:E1026–E1034. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Sui Y, Sun M, Wu F, Yang L, Di W, Zhang G, Zhong L, Ma Z, Zheng J, Fang X and Ma T: Inhibition of TMEM16A expression suppresses growth and invasion in human colorectal cancer cells. PLoS One. 9:e1154432014. View Article : Google Scholar : PubMed/NCBI

97 

Cao Q, Liu F, Ji K, Liu N, He Y, Zhang W and Wang L: MicroRNA-381 inhibits the metastasis of gastric cancer by targeting TMEM16A expression. J Exp Clin Cancer Res. 36:292017. View Article : Google Scholar : PubMed/NCBI

98 

Lee YS, Lee JK, Bae Y, Lee BS, Kim E, Cho CH, Ryoo K, Yoo J, Kim CH, Yi GS, et al: Suppression of 14-3-3γ-mediated surface expression of ANO1 inhibits cancer progression of glioblastoma cells. Sci Rep. 6:264132016. View Article : Google Scholar

99 

Shang L, Hao JJ, Zhao XK, He JZ, Shi ZZ, Liu HJ, Wu LF, Jiang YY, Shi F, Yang H, et al: ANO1 protein as a potential biomarker for esophageal cancer prognosis and precancerous lesion development prediction. Oncotarget. 7:24374–24382. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Akpalo H, Lange C and Zustin J: Discovered on gastrointestinal stromal tumour 1 (DOG1): A useful immunohistochemical marker for diagnosing chondroblastoma. Histopathology. 60:1099–1106. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Chang Z, Cai C, Han D, Gao Y, Li Q, Feng L, Zhang W, Zheng J, Jin J, Zhang H and Wei Q: Anoctamin5 regulates cell migration and invasion in thyroid cancer. Int J Oncol. 51:1311–1319. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Li Y, Wang X, Vural S, Mishra NK, Cowan KH and Guda C: Exome analysis reveals differentially mutated gene signatures of stage, grade and subtype in breast cancers. PLoS One. 10:e01193832015. View Article : Google Scholar : PubMed/NCBI

103 

Chen W, Gu M, Gao C, Chen B, Yang J, Xie X, Wang X, Sun J and Wang J: The prognostic value and mechanisms of TMEM16A in human cancer. Front Mol Biosci. 8:5421562021. View Article : Google Scholar : PubMed/NCBI

104 

Liu F, Yang X, Geng M and Huang M: Targeting ERK, an Achilles' heel of the MAPK pathway, in cancer therapy. Acta Pharm Sin B. 8:552–562. 2018. View Article : Google Scholar : PubMed/NCBI

105 

Wang H, Yao F, Luo S, Ma K, Liu M, Bai L, Chen S, Song C, Wang T, Du Q, et al: A mutual activation loop between the Ca2+-activated chloride channel TMEM16A and EGFR/STAT3 signaling promotes breast cancer tumorigenesis. Cancer Lett. 455:48–59. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Bai W, Liu M and Xiao Q: The diverse roles of TMEM16A Ca2+-activated Cl-channels in inflammation. J Adv Res. 33:53–68. 2021. View Article : Google Scholar : PubMed/NCBI

107 

Lin Z, Deng Z, Liu J, Lin Z, Chen S, Deng Z and Li W: Chloride channel and inflammation-mediated pathogenesis of osteoarthritis. J Inflamm Res. 15:953–964. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Liu J, Liu Y, Ren Y, Kang L and Zhang L: Transmembrane protein with unknown function 16A overexpression promotes glioma formation through the nuclear factor-κB signaling pathway. Mol Med Report. 9:1068–1074. 2014. View Article : Google Scholar

109 

Zhou L, Deng ZZ, Li HY, Jiang N, Wei ZS, Hong MF, Chen XD, Wang JH, Zhang MX, Shi YH, et al: TRIM31 promotes glioma proliferation and invasion through activating NF-κB pathway. Onco Targets Ther. 12:2289–2297. 2019. View Article : Google Scholar :

110 

Duvvuri U, Shiwarski DJ, Xiao D, Bertrand C, Huang X, Edinger RS, Rock JR, Harfe BD, Henson BJ, Kunzelmann K, et al: TMEM 16 A induces MAPK and contributes directly to tumorigenesis and cancer progression. Cancer Res. 72:3270–3281. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Deng L, Yang J, Chen H, Ma B, Pan K, Su C, Xu F and Zhang J: Knockdown of TMEM16A suppressed MAPK and inhibited cell proliferation and migration in hepatocellular carcinoma. Onco Targets Ther. 9:325–333. 2016.PubMed/NCBI

112 

Ruiz C, Martins JR, Rudin F, Schneider S, Dietsche T, Fischer CA, Tornillo L, Terracciano LM, Schreiber R, Bubendorf L and Kunzelmann K: Enhanced expression of ANO1 in head and neck squamous cell carcinoma causes cell migration and correlates with poor prognosis. PLoS One. 7:e432652012. View Article : Google Scholar : PubMed/NCBI

113 

Dixit R, Kemp C, Kulich S, Seethala R, Chiosea S, Ling S, Ha PK and Duvvuri U: TMEM16A/ANO1 is differentially expressed in HPV-negative versus HPV-positive head and neck squamous cell carcinoma through promoter methylation. Sci Rep. 5:166572015. View Article : Google Scholar : PubMed/NCBI

114 

Mokutani Y, Uemura M, Munakata K, Okuzaki D, Haraguchi N, Takahashi H, Nishimura J, Hata T, Murata K, Takemasa I, et al: Down-regulation of microRNA-132 is associated with poor prognosis of colorectal cancer. Ann Surg Oncol. 23:599–608. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Lin S and Gregory RI: MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Wang H, Zou L, Ma K, Yu J, Wu H, Wei M and Xiao Q: Cell-specific mechanisms of TMEM16A Ca2+-activated chloride channel in cancer. Mol Cancer. 16:1522017. View Article : Google Scholar

117 

Wanitchakool P, Wolf L, Koehl GE, Sirianant L, Schreiber R, Kulkarni S, Duvvuri U and Kunzelmann K: Role of anoctamins in cancer and apoptosis. Philos Trans R Soc Lond B Biol Sci. 369:201300962014. View Article : Google Scholar : PubMed/NCBI

118 

Ahn SY, Yang JH, Kim NH, Lee K, Cha YH, Yun JS, Kang HE, Lee Y, Choi J, Kim HS and Yook J: Anti-helminthic niclosamide inhibits Ras-driven oncogenic transformation via activation of GSK-3. Oncotarget. 8:31856–31863. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Miner K, Labitzke K, Liu B, Wang P, Henckels K, Gaida K, Elliott R, Chen JJ, Liu L, Leith A, et al: Drug repurposing: The anthelmintics niclosamide and nitazoxanide are potent TMEM16A antagonists that fully bronchodilate airways. Front Pharmacol. 10:512019. View Article : Google Scholar : PubMed/NCBI

120 

Jin Y, Lu Z, Ding K, Li J, Du X, Chen C, Sun X, Wu Y, Zhou J and Pan J: Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: Inactivation of the NF-κB pathway and generation of reactive oxygen species. Cancer Res. 70:2516–2527. 2010. View Article : Google Scholar : PubMed/NCBI

121 

Han Z, Li Q, Wang Y, Wang L, Li X, Ge N, Wang Y and Guo C: Niclosamide induces cell cycle arrest in G1 phase in head and neck squamous cell carcinoma through Let-7d/CDC34 Axis. Front Pharmacol. 9:15442019. View Article : Google Scholar : PubMed/NCBI

122 

Li Y, Li PK, Roberts MJ, Arend RC, Samant RS and Buchsbaum DJ: Multi-targeted therapy of cancer by niclosamide: A new application for an old drug. Cancer Lett. 349:8–14. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Arend RC, Londoño-Joshi AI, Gangrade A, Katre AA, Kurpad C, Li Y, Samant RS, Li PK, Landen CN, Yang ES, et al: Correction: Niclosamide and its analogs are potent inhibitors of Wnt/β-catenin, mTOR and STAT3 signaling in ovarian cancer. Oncotarget. 9:19459. 2018. View Article : Google Scholar

124 

Lafkas D, Shelton A, Chiu C, de Leon Boenig G, Chen Y, Stawicki SS, Siltanen C, Reichelt M, Zhou M, Wu X, et al: Therapeutic antibodies reveal Notch control of transdifferentiation in the adult lung. Nature. 528:127–131. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Danahay H, Pessotti AD, Coote J, Montgomery BE, Xia D, Wilson A, Yang H, Wang Z, Bevan L, Thomas C, et al: Notch2 is required for inflammatory cytokine-driven goblet cell metaplasia in the lung. Cell Rep. 10:239–252. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Seo Y, Kim J, Chang J, Kim SS, Namkung W and Kim I: Synthesis and biological evaluation of novel Ani9 derivatives as potent and selective ANO1 inhibitors. Eur J Medicinal Chem. 160:245–255. 2018. View Article : Google Scholar

127 

Burock S, Daum S, Keilholz U, Neumann K, Walther W and Stein U: Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: The NIKOLO trial. BMC Cancer. 18:2972018. View Article : Google Scholar : PubMed/NCBI

128 

Schweizer MT, Haugk K, McKiernan JS, Gulati R, Cheng HH, Maes JL, Dumpit RF, Nelson PS, Montgomery B, McCune JS, et al: Correction: A phase I study of niclosamide in combination with enzalutamide in men with castration-resistant prostate cancer. PLoS One. 13:e02027092018. View Article : Google Scholar : PubMed/NCBI

129 

Yan Y, Ding X, Han C, Gao J, Liu Z, Liu Y and Wang K: Involvement of TMEM16A/ANO1 upregulation in the oncogenesis of colorectal cancer. Biochim Biophys Acta Mol Basis Dis. 1868:1663702022. View Article : Google Scholar : PubMed/NCBI

130 

Khalil A, Elesawy BH, Ali TM and Ahmed OM: Bee venom: From venom to drug. Molecules. 26:49412021. View Article : Google Scholar : PubMed/NCBI

131 

Badawi JK: Bee venom components as therapeutic tools against prostate cancer. Toxins (Basel). 13:3372021. View Article : Google Scholar : PubMed/NCBI

132 

Schreiber R, Ousingsawat J, Wanitchakool P, Sirianant L, Benedetto R, Reiss K and Kunzelmann K: Regulation of TMEM16A/ANO1 and TMEM16F/ANO6 ion currents and phospholipid scrambling by Ca2+ and plasma membrane lipid. J Physiol. 596:217–229. 2017. View Article : Google Scholar

133 

Simões F, Ousingsawat J, Wanitchakool P, Fonseca A, Cabrita I, Benedetto R, Schreiber R and Kunzelmann K: CFTR supports cell death through ROS-dependent activation of TMEM16F (anoctamin 6). Pflugers Arch. 470:305–314. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Xue Q, Yan D, Chen X, Li X, Kang R, Klionsky DJ, Kroemer G, Chen X, Tang D and Liu J: Copper-dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy. 19:1982–1996. 2023. View Article : Google Scholar : PubMed/NCBI

136 

El-Didamony SE, Amer RI and El-Osaily GH: Formulation, characterization and cellular toxicity assessment of a novel bee-venom microsphere in prostate cancer treatment. Sci Rep. 12:132132022. View Article : Google Scholar : PubMed/NCBI

137 

Schreiber R, Buchholz B, Kraus A, Schley G, Scholz J, Ousingsawat J and Kunzelmann K: Lipid peroxidation drives renal cyst growth in vitro through activation of TMEM16A. J Am Soc Nephrol. 30:228–242. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Schmaier AA, Anderson PF, Chen SM, El-Darzi E, Aivasovsky I, Kaushik MP, Sack KD, Hartzell HC, Parikh SM, Flaumenhaft R and Schulman S: TMEM16E regulates endothelial cell procoagulant activity and thrombosis. J Clin Invest. 133:e1638082023. View Article : Google Scholar : PubMed/NCBI

139 

Fujii Y, Taniguchi M, Nagaya S, Ueda Y, Hashizume C, Watanabe K, Takeya H, Kosaka T and Okazaki T: A novel mechanism of thrombocytopenia by PS exposure through TMEM16F in sphingomyelin synthase 1 deficiency. Blood Adv. 5:4265–4277. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Filep JG: Two to tango: Endothelial cell TMEM16 scramblases drive coagulation and thrombosis. J Clin Invest. 133:e1706432023. View Article : Google Scholar : PubMed/NCBI

141 

Wu N, Cernysiov V, Davidson D, Song H, Tang J, Luo S, Lu Y, Qian J, Gyurova IE, Waggoner SN, et al: Critical role of lipid scramblase TMEM16F in phosphatidylserine exposure and repair of plasma membrane after pore formation. Cell Rep. 30:1129–1140.e5. 2020. View Article : Google Scholar : PubMed/NCBI

142 

Taylor KA and Mahaut-Smith MP: A major interspecies difference in the ionic selectivity of megakaryocyte Ca2+-activated channels sensitive to the TMEM16F inhibitor CaCCinh-A01. Platelets. 30:962–966. 2019. View Article : Google Scholar :

143 

Yang H, Kim A, David T, Palmer D, Jin T, Tien J, Huang F, Cheng T, Coughlin SR, Jan YN and Jan LY: TMEM16F Forms a Ca2+-activated cation channel required for lipid scrambling in platelets during blood coagulation. Cell. 151:111–122. 2012. View Article : Google Scholar : PubMed/NCBI

144 

Yu H, Wang Z, Li Z, An Y, Yan M, Ji S, Xu M, Wang L, Dong W, Shi J and Gao C: Hyperuricemia enhances procoagulant activity of vascular endothelial cells through TMEM16F regulated phosphatidylserine exposure and microparticle release. FASEB J. 35:e218082021. View Article : Google Scholar : PubMed/NCBI

145 

Goyal P, Choi JJ, Pinheiro LC, Schenck EJ, Chen R, Jabri A, Satlin MJ, Campion TR Jr, Nahid M, Ringel JB, et al: Clinical characteristics of Covid-19 in new york city. N Engl J Med. 382:2372–2374. 2020. View Article : Google Scholar : PubMed/NCBI

146 

Levi M, Thachil J, Iba T and Levy JH: Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 7:e438–e440. 2020. View Article : Google Scholar : PubMed/NCBI

147 

Edler C, Schröder AS, Aepfelbacher M, Fitzek A, Heinemann A, Heinrich F, Klein A, Langenwalder F, Lütgehetmann M, Meißner K, et al: Correction to: Dying with SARS-CoV-2 infection-an autopsy study of the first consecutive 80 cases in Hamburg, Germany. Int J Legal Med. 134:19772020. View Article : Google Scholar : PubMed/NCBI

148 

Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 181:271–280.e8. 2020. View Article : Google Scholar : PubMed/NCBI

149 

Hoffmann M, Hofmann-Winkler H and Pöhlmann S: Priming time: How Cellular Proteases Arm Coronavirus Spike Proteins. Springer International Publishing; Cham: pp. 71–98. 2018

150 

Braga L, Ali H, Secco I, Chiavacci E, Neves G, Goldhill D, Penn R, Jimenez-Guardeño JM, Ortega-Prieto AM, Bussani R, et al: Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature. 594:88–93. 2021. View Article : Google Scholar : PubMed/NCBI

151 

Cappelletto A, Allan HE, Crescente M, Schneider E, Bussani R, Ali H, Secco I, Vodret S, Simeone R, Mascaretti L, et al: SARS-CoV-2 Spike protein activates TMEM16F-mediated platelet procoagulant activity. Front Cardiovasc Med. 9:10132622023. View Article : Google Scholar : PubMed/NCBI

152 

Abdulamir AS, Gorial FI, Saadi SJ, Maulood MF, Hashim HA, Alnuaimi AS and Abdulrrazaq MK: A randomised controlled trial of effectiveness and safety of Niclosamide as add on therapy to the standard of care measures in COVID-19 management. Ann Med Surg (Lond). 69:1027792021.PubMed/NCBI

153 

Chen W, Mook RA Jr, Premont RT and Wang J: Niclosamide: Beyond an antihelminthic drug. Cell Signal. 41:89–96. 2018. View Article : Google Scholar

154 

Kalienkova V, Clerico Mosina V and Paulino C: The Groovy TMEM16 family: Molecular mechanisms of lipid scrambling and ion conduction. J Mol Biol. 433:1669412021. View Article : Google Scholar : PubMed/NCBI

155 

Lam AKM, Rutz S and Dutzler R: Inhibition mechanism of the chloride channel TMEM16A by the pore blocker 1PBC. Nat Commun. 13:27982022. View Article : Google Scholar : PubMed/NCBI

156 

Bill A, Gutierrez A, Kulkarni S, Kemp C, Bonenfant D, Voshol H, Duvvuri U and Gaither LA: ANO1/TMEM16A interacts with EGFR and correlates with sensitivity to EGFR-targeting therapy in head and neck cancer. Oncotarget. 6:9173–9188. 2015. View Article : Google Scholar : PubMed/NCBI

157 

Zhang X, Zhang G, Zhai W, Zhao Z, Wang S and Yi J: Inhibition of TMEM16A Ca2+-activated Cl-channels by avermectins is essential for their anticancer effects. Pharmacol Res. 156:1047632020. View Article : Google Scholar

158 

Fang H, Deng Z, Liu J, Chen S, Deng Z and Li W: The mechanism of bone remodeling after bone aging. Clin Interv Aging. 17:405–415. 2022. View Article : Google Scholar : PubMed/NCBI

159 

Genovese M, Buccirossi M, Guidone D, De Cegli R, Sarnataro S, di Bernardo D and Galietta LJV: Analysis of inhibitors of the anoctamin-1 chloride channel (transmembrane member 16A, TMEM16A) reveals indirect mechanisms involving alterations in calcium signalling. Br J Pharmacol. 180:775–785. 2023. View Article : Google Scholar

160 

Shaibani A, Khan S and Shinawi M: Autosomal dominant ANO5-Related disorder associated with myopathy and gnathodiaphyseal dysplasia. Neurol Genet. 7:e6122021. View Article : Google Scholar : PubMed/NCBI

161 

Liu X, Wang X, Ma X, Li H, Miao C, Tian Z and Hu Y: Genetic disruption of Ano5 leads to impaired osteoclastogenesis for gnathodiaphyseal dysplasia. Oral Dis. 30:1403–1415. 2024. View Article : Google Scholar

162 

Chandra G, Defour A, Mamchoui K, Pandey K, Mishra S, Mouly V, Sreetama S, Mahad Ahmad M, Mahjneh I, Morizono H, et al: Dysregulated calcium homeostasis prevents plasma membrane repair in Anoctamin 5/TMEM16E-deficient patient muscle cells. Cell Death Discov. 5:1182019. View Article : Google Scholar : PubMed/NCBI

163 

Thiruvengadam G, Sreetama SC, Charton K, Hogarth M, Novak JS, Suel-Petat L, Chandra G, Allard B, Richard I and Jaiswal JK: Anoctamin 5 Knockout mouse model recapitulates LGMD2L muscle pathology and offers insight into in vivo functional deficits. J Neuromuscul Dis. 8(Suppl): S243–S255. 2021. View Article : Google Scholar : PubMed/NCBI

164 

Li H, Wang X, Chen E, Liu X, Ma X, Miao C, Tian Z, Dong R and Hu Y: Introduction of a Cys360Tyr Mutation in ANO5 creates a mouse model for gnathodiaphyseal dysplasia. J Bone Miner Res. 37:515–530. 2022. View Article : Google Scholar

165 

Jiang LT, Li LX, Liu Y, Zhang XL, Pan YG, Wang L, Wan XH and Jin LJ: The expanding clinical and genetic spectrum of ANO3 dystonia. Neurosci Lett. 746:1355902021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 54 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang Z, Iqbal Z, Zhao Z, Chen X, Mahmmod A, Liu J, Li W and Deng Z: TMEM16 proteins: Ca<sup>2+</sup>‑activated chloride channels and phospholipid scramblases as potential drug targets (Review). Int J Mol Med 54: 81, 2024.
APA
Huang, Z., Iqbal, Z., Zhao, Z., Chen, X., Mahmmod, A., Liu, J. ... Deng, Z. (2024). TMEM16 proteins: Ca<sup>2+</sup>‑activated chloride channels and phospholipid scramblases as potential drug targets (Review). International Journal of Molecular Medicine, 54, 81. https://doi.org/10.3892/ijmm.2024.5405
MLA
Huang, Z., Iqbal, Z., Zhao, Z., Chen, X., Mahmmod, A., Liu, J., Li, W., Deng, Z."TMEM16 proteins: Ca<sup>2+</sup>‑activated chloride channels and phospholipid scramblases as potential drug targets (Review)". International Journal of Molecular Medicine 54.4 (2024): 81.
Chicago
Huang, Z., Iqbal, Z., Zhao, Z., Chen, X., Mahmmod, A., Liu, J., Li, W., Deng, Z."TMEM16 proteins: Ca<sup>2+</sup>‑activated chloride channels and phospholipid scramblases as potential drug targets (Review)". International Journal of Molecular Medicine 54, no. 4 (2024): 81. https://doi.org/10.3892/ijmm.2024.5405