Open Access

The roles and functions of TMEM protein family members in cancers, cardiovascular and kidney diseases (Review)

  • Authors:
    • Haosen Xu
    • Shanzhi Yang
    • Peimin Liu
    • Yan Zhang
    • Ting Zhang
    • Jinyi Lan
    • Huan Jiang
    • Danfeng Wu
    • Jiaoqing Li
    • Xiaoyan Bai
  • View Affiliations

  • Published online on: February 11, 2025     https://doi.org/10.3892/br.2025.1941
  • Article Number: 63
  • Copyright: © Xu 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

Transmembrane protein (TMEM) is a type of membrane proteins, encoded by TMEM gene, also known as integral membrane protein. TMEM gene family contains various members and its encoded proteins have various functions and expressed in numerous organs. It has been proved to be widely involved in the formation of a lot of organelle membranes, enzymes, receptors and channels, mediating numerous normal physiological functions and regulating various disease processes. At present, accumulating evidences at home and abroad have shown that TMEM is involved in regulating the occurrence and development of different tumors, cardiovascular and kidney diseases. The improved understanding of molecular mechanisms of TMEM genes and proteins may provide new directions and ideas for the prevention, diagnosis and treatment of diseases. In the present review, the roles of TMEM and biological functions in various cancers, cardiovascular and kidney diseases were discussed.

1. Introduction

Membrane proteins, ubiquitously distributed both on the cellular and organelle membranes across multiple cell types in the body, serve as pivotal mediators of information exchange, material transition, and signal transduction among cells and their interior milieu (1); thus, they are referred to as the principal custodians of biofilm function. The categorization of these proteins spans lipid-anchored membrane proteins, integrated membrane proteins and peripheral membrane proteins, based on the complexities inherent in their separation and their spatial localization within the membrane (2). Notably, integrated membrane proteins, also designated as transmembrane (TMEM) proteins, constitute a substantial majority (70-80%). TMEM proteins include extracellular region, transmembrane region and cytoplasmic region, with at least one or more hydrophobic transmembrane domains inserted into the lipid bilayer (3).

The TMEM gene family, along with its myriad encoded proteins, as validated by prior research, plays significant roles in formulating various organelle membranes, including mitochondria (4,5), endoplasmic reticulum (6,7), Golgi apparatus (8) and lysosomes (9,10). Therefore, TMEM proteins could exert various biological functions. As such, TMEM proteins exhibit a broad spectrum of biological functionality including their roles as diverse receptors (11,12), enzymes (13) and channels (14). Furthermore, by activating or inhibiting various signaling pathways, TMEM proteins are found to be indispensable in physiological processes as well as in the initiation and progression of myriad diseases. Amongst the TMEM family, TMEM16A is the most extensively studied member, and correlations between TMEM and disease pathogenesis have been systematically and comprehensively examined in conditions such as tumors, cardiovascular disorders and kidney diseases. The present review aims to make a systematic and comprehensive summary of TMEM and the aforementioned three aspects.

2. TMEM and cancers

TMEM family members ubiquitously exist across different systems and tissues, and are closely related to various diseases. Their roles in the onset and progression of malignant tumors have been studied widely. Through the activation of diverse signaling pathways, TMEM family plays a critical role in pro-tumor, antitumor and the heterogeneity of tumors. Several recent investigations have elaborated the relationship between TMEM and cancers (Table I).

Table I

The roles and functions of TMEMs in various cancers.

Table I

The roles and functions of TMEMs in various cancers.

First author/s, yearTMEMDisease typesAssociated tissues or cellsAlterationsPathwayAssociated targetsFunction or mechanism(Refs.)
Fu et al, 2024TMEM205GCGES-1, SGC-7901UpregulationWnt/β-cateninc-mycPromote proliferation, stemness, EMT, migration and angiogenesis(15)
Guo et al, 2010TMEM8B (NGX6)Colon cancerHT-29DownregulationWnt/β-cateninTCF-4, c-myc, cyclin D1 and COX-2Inhibit cell invasion and adhesion(16)
Chen et al, 2023; Liu et al, 2019; Liu et al, 2015TMEM196Lung cancerHBE, 293T SPC-A-1, A549 and LTEP-a-2,DownregulationWnt/β-cateninMMP2 and MMP7; p21, Bax, cyclin D1, c-myc, CD44 and β-cateninInhibit metastasis and progression(17,48,49)
Ng et al, 2014TMEM98HCCMHCC97L, PLC, Hep3BUpregulationAKTAKT, GSK3-β, BCL-XL, P53Confer chemoresistance(18)
Zhang et al, 2021TMEM229ANSCLCBEAS-2B, A549, H23, H226, 95D, H1975, PC-9 and H1299DownregulationAKT/ERKAKT, ERK, MMP2, Snail 1, E-cadherin, N-cadherinSuppress cell EMT, proliferation, migration and invasion(19)
Jun et al, 2017TMEM16J (ANO9)Pancreatic cancerhTERT-HPNE, PANC-1, AsPC-1, BxPC-3 and Capan-2,UpregulationMAPKERK1/2, EGFRIncrease cell proliferation(20)
Zhao et al, 2018TMEM17Breast cancerMCF-10A, MCF-7, T47D, MDA-MB-231 and MDA-MB-468UpregulationAKT/GSK3ββ-catenin, Snail, c-myc and cyclin D1 and E-cadherinPromote malignant progression(32)
Wang et al, 2024 GlioblastomaLN229 and U87MGUpregulationPI3K/AKTYY1, PI3K, AKTPromote cell proliferation, migration, and invasion, reduce apoptosis,(33)
Zhang et al, 2017 NSCLCHBE, A549, H292, H1299, Calu-1, SK-MES-1, SPC-A-1 and LK2DownregulationMAPKERK, P90RSK, Snail, Occludin and Zo-1Inhibit cell invasion and migration(34)
Flamant et al, 2012TMEM45ABreast and liver cancerMDA-MB-231, HepG2UpregulationN/Acaspase 3Promote hypoxia-induced chemoresistance(35)
Guo et al, 2015 Ovarian cancerOVCAR3, A2780, HO-8910, CAOV3, SK-OV-3 and 293TUpregulationTGF-β, WntTGF-β1, TGF-β2, RhoA and ROCK2Promote cell proliferation, adhesion and invasion(36)
Shen et al, 2018TMEM45BGCBGC-823, MGC-803, SGC-7901 and HGC-27UpregulationJAK2/STAT3JAK2, STAT3Promote cell proliferation, migration and invasion(37)
Li et al, 2017 OsteosarcomaU2OS, SaOS2, MG-63 and hFOB1.19UpregulationWnt/β-cateninβ-catenin, cyclin D1, and c-mycPromote cell proliferation, migration, and invasion(38)
Zhang et al, 2015TMEM88NSCLCHBE, A549, H1299, H460, H292, SPC-A-1 (SPC), LTEP-A-2, PG-BE1 and PG-LH7UpregulationWntDVLS, p38, GSK3β (Thr390), Snail, Zo-1, OccludinPromote invasion and metastasis(39)
Yu et al, 2015 Breast cancerMCF-10A, MCF-7, HER18, MDA-MB-231, and MDA-MB-468UpregulationWntDVLS, Snail, Zo-1, OccludinPromote invasion and metastasis(40)
De Leon et al, 2016 Ovarian cancerA2780, CP70, PEO1 and PEO4UpregulationWntPITX2, FOSL1, c-Myc, Cyclin D1Promoter hypomethylation promote cancer cell platinum resistance(41)
Cheng et al, 2015TMEM158Ovarian cancerOVCAR3, A2780, HO-8910, CAOV3, SK-OV-3 and 293TUpregulationTGF-βTGF-β1, BMP4, ICAM-1 and VCAM-1,Promote cell proliferation, adhesion and invasion(42)
Fu et al, 2020 Pancreatic cancerSW1990, COLO357, MIAPACA-2, PANC-1, CFPAC-1, and BXPC-3UpregulationTGF-β and PI3K/AKTE-cadherin, Vimentin, N-cadherin, and α-SMA, MMP-2/9Promote cell proliferation, migration, and invasion(43)
Xu et al, 2014TMEM97 (MAC30)GCBGC-823, AGSUpregulationAKTcyclin B1 and WAVE2Promote cell proliferation migration, and invasion(44)
Zhu et al, 2021 Breast cancerMCF-10A, MCF7, BT549, Bcap37, and MDA-MB-231UpregulationWnt/β-catenin and PI3K/AktE-cadherin, N-cadherin, vimentin, Slug, Snail, Twist, ZEB1 AKT, β-catenin, survivin, and cyclin D1Promote cell invasion and EMT(45)
Mao et al, 2022 Colorectal cancerSW480, No. iCell-h204; SW620, No. iCell-h206; LoVo, No. iCell-h126; HCT116, No. iCell-h071 and DLD1, No. iCell-h053Upregulation GSK-3β/β-cateninYY1, GSK-3β/β-catenin, PCNA, cleaved caspase 8/3/7 and cleaved PARPInhibit cell invasion migration, growth and enhance apoptosi(46)
Xu et al, 2024 CRCFHC, HCT15, HCT8, HCT116, and SW480UpregulationAKT/mTORATP-binding cassette, E-cadherin, Vimentin, N-cadherin, P-glycoprotein, ABCC1, ABCC2Promote 5-FU resistance(47)

[i] TMEM, transmembrane; NSCLC, non-small cell lung cancer; GC, gastric cancer; HBE cells, human normal bronchial epithelial cells; FHC, normal colorectal mucosa cells; CRC, colorectal cancer; N/A, not available.

Wnt and RTK related signaling pathways

TMEM205 overexpresses in cisplatin-resistant gastric cancer (GC), which promotes proliferation, stemness, epithelial-mesenchymal transition (EMT), migration and angiogenesis of GC cells via activation of the Wnt/β-catenin signaling pathway (15). In colon cancer cells, the expression of TMEM8B decreases. TMEM8B could limit the translocation of beta-catenin from nucleus and cytoplasm to plasma membrane, and inhibit the adhesion and invasion of colon cancer cells by suppressing the transcription factor TCF-4 and downregulating the expression of downstream target genes c-Myc, cyclin D1 and COX-2 in the Wnt signaling pathway (16). The expression levels of TMEM196 are significantly decreased in lung cancer tissues and cells, and its low expression in clinical patients is associated with poor prognosis. TMEM196 can inhibit the Wnt signaling pathway and repress β-catenin promoter transcription in vitro and in vivo. TMEM196 silencing results in significant upregulation of the expression of downstream target genes MMP2 and MMP7, attenuating the anti-metastatic effect (17).

In chemoresistant hepatocellular carcinoma (HCC) cells, the silence/overexpression of TMEM98 would restore/reduce their chemosensitivity, in which forced overexpression of TMEM98 may inhibit the expression of p53 via activating the AKT signaling pathway, thereby fostering drug resistance in tumor cells. Therefore, TMEM98 functions as a chemoresistance-conferring gene in HCC (18). The expression levels of TMEM229A are significantly decreased in non-small cell lung cancer (NSCLC). Survival analysis identifies that low expression of TMEM229A was associated with a poor prognosis. TMEM229A overexpression would suppress the EMT effectively, increasing E-cadherin expression and reducing N-cadherin, Snail family transcriptional repressor 1 and MMP2 expression, thereby significantly inhibiting cell proliferation, migration and invasion. Additionally, TMEM229A overexpression reduces the expression levels of phosphorylated (p)-ERK and p-AKT, which can be partially suppressed by the specific ERK inhibitor PD98059(19).

TMEM16J is highly expressed in pancreatic cancer, accompanied with the activation of the MAPK signaling pathway. TMEM16J forms a protein complex binding to EGFR, which will accelerate the proliferation of cancer cells, thereby deteriorating the patients' prognosis and survival rates. However, specific knockdown of TMEM16J could inhibit the phosphorylation of ERK1/2 and EGFR, and reduce the expression level of total protein, thereby inhibiting the proliferation activity of cancer cells, and enhancing the anticancer effect of drugs such as gemcitabine and erlotinib (20). Hence, TMEM16J appears to be a potential therapeutic target and clinically prognostic marker for pancreatic cancer.

Multiple signaling pathways

TMEM16A can affect the growth of different tumors by activating various signaling pathways, including MAPK or EGFR signaling in colorectal cancer (CRC) (21), HCC (22) and head and neck squamous cell carcinoma (23,24), EGFR and Wnt/CAMKII signaling in breast cancer (25), TGF-β signaling in GC (26) and NF-κB signaling in glioma (27). Furthermore, TMEM16A is also associated with lung cancer (28), pancreatic cancer (29), prostate cancer (30) and esophageal squamous cell carcinoma (31), but the specific mechanism require further elucidation.

The expression level of TMEM17 is upregulated in breast cancer tissues, which upregulates active β-catenin, Snail and downregulates E-cadherin through the AKT/GSK3β signaling pathway, enhancing the proliferation, invasion and migration of breast cancer cells. However, these effects reversed by AKT inhibitor LY294002 in vitro (32). YY1 (a well-recognized oncogenic transcription factor) increases the expression of TMEM17 in glioblastoma, which promotes the proliferation, migration and invasion of cancer cells through the AKT/PI3K pathway; and reduces apoptosis, ultimately leading to the high malignancy of glioblastoma (33). The PI3K activator 740Y-P can reverse the effects caused by TMEM17 knockdown. However, the expression level of TMEM17 in NSCLC is significantly lower than adjacent normal lung tissues. Overexpression of TMEM17 downregulates the levels of p-ERK and its downstream molecules p-P90RSK and Snail while the levels of Occludin and Zo-1 are upregulated, inhibiting the invasive and migratory ability of lung cancer cells. Therefore, TMEM17 could be a negative regulator in NSCLC (34).

Elevated expression of TMEM45A has been observed in liver cancer and breast cancer, and its overexpression will induce the chemoresistance of tumor cells under hypoxia. Thus, the availability of TMEM45A potentially renders it a biomarker of chemotherapy resistance (35). Notably, overexpression of TMEM45A not only features in liver and breast cancer, but also in ovarian cancer cells. Conversely, knockdown of TMEM45A can markedly downregulate the expression of TGF-β1, TGF-β2, RhoA and ROCK2 so as to markedly restrain tumor cell proliferation, adhesion and invasion. This discovery reveals that TMEM45A participates in regulating TGF-β and Wnt signaling pathways, offering insights towards potential therapeutic targets (36). TMEM45B is highly expressed in GC tissue cells in patients. Knockdown of TMEM45B can inhibit the proliferation, migration, invasion and EMT of GC cells via suppressing the expression of p-JAK2 and p-STAT3 through the JAK2/STAT3 signaling pathway in cancer cells. Therefore, TMEM45B may become a new therapeutic molecular target for GC (37). The expression of TMEM45B is obviously upregulated in human osteosarcoma U2OS cell lines, knockdown of TMEM45B can significantly suppress the expression of beta-catenin, cyclin D1 and c-Myc, and inhibit the proliferation, migration and invasion of U2OS cells (38).

TMEM88 is highly expressed in NSCLC (39) and triple negative breast cancer tissues (40). It enhances invasive and metastatic cell characters via promoting Snail expression and inhibiting Zo-1 and Occludin expression, which is associated to Dishevelled of the Wnt signaling pathway. In addition, the expression of TMEM88 is increased due to promoter hypomethylation in ovarian cancer, which is closely related to platinum resistance of tumor cells. However, knockdown of TMEM88 leads to notable upregulation of downstream genes c-Myc and cyclin-D1 of the Wnt signaling pathway, which can promote the proliferation and colony formation of cancer cells, and also re-sensitize cells to cisplatin (41).

The expression of TMEM158 in ovarian and pancreatic cancer is increased. Knockdown of TMEM158 would significantly inhibit the proliferation of ovarian cancer cells by curbing the TGF-β signaling pathway, and also obviously inhibit cell adhesion by downregulating the expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) (42). TMEM158 enhances the proliferation, migration and invasion of pancreatic cancer cells by activating TGF-β1 and PI3K/AKT signaling pathways to accelerate cell cycle, EMT and MMP-2/9 productions (43).

TMEM97 is a conserved integral membrane protein highly expressed in various human cancers including GC, breast cancer and CRC. Upregulation of TMEM97 has been associated with progression and poor outcome in cancers. Knockdown of TMEM97 in GC cells can restrain the degree of phosphorylation of AKT and reduce the expression level of cyclin B1 and WAVE2, leading to inhibit proliferation and mobility of GC cells (44). In breast cancer cells, TMEM97 deficiency attenuates the Wnt/β-catenin signaling cascade via regulating LRP6 phosphorylation, leading to a decrease in the expression of Wnt target genes AXIN2, LEF1 and survivin. Meanwhile, knockdown of TMEM97 suppresses cell viability, proliferation, colony formation, migration, invasion and stemness properties in breast cancer cells. Importantly, TMEM97 deficiency was revealed to suppress tumor growth through downregulating the Wnt/β-catenin signaling pathway in breast cancer xenograft mouse models (45). In CRC, TMEM97 is transcriptionally activated by YY1 and promotes CRC progression via the GSK-3β/β-catenin signaling pathway. The silencing of TMEM97 inhibits invasion and migration of CRC cells in vitro and leads to suppress growth and enhance apoptosis in cells and xenografts (46). Another study demonstrated that TMEM97 knockdown suppresses EMT, expression of ATP-binding cassette transporters via inactivating the AKT/mTOR pathway, thus attenuating 5-FU resistance in CRC cells (47). These findings demonstrate that TMEM97-targeted inhibition may be a promising therapeutic strategy for cancer.

There are numerous members in TMEM family, most of which are involved in the occurrence and development of various tumors, performing an indispensable role in the proliferation, migration and invasion of diverse tumor cells. Most of TMEM proteins are increased in tumors: TMEM17 in glioblastoma (33), TMEM45A in breast and liver cancer (35) and ovarian cancer (36); TMEM45B in GC (37) and osteosarcoma (38); TMEM88 in NSCLC (39), breast (40) and ovarian cancer (41); TMEM158 in ovarian (42) and pancreatic cancer (43); TMEM97 in GC (44), breast (45) and colon cancer (46). They promote proliferation, invasion and metastasis of tumor cells. Among these TMEM proteins whose expression is upregulated, TMEM98(18), TMEM45A (35) and TMEM88(41) promote chemotherapy resistance in different ways. If there are inhibitors targeting these TMEM proteins, it may restore chemotherapy sensitivity of cancer cells and improve patients' prognosis and survival time. On the contrary, a small amount of TMEM members, TMEM8B (16), TMEM196(48) and TMEM229A (19), have inhibitory effects on the occurrence and development of tumors, and their expression levels are downregulated. Notably, the role of TMEM196 in lung cancer was demonstrated in genetic model animals. The hypermethylation and low expression level of TMEM196 are significantly associated with age, worse pathological type and poorer survival of patients, thus it is considered a biomarker of lung cancer in the clinic (48,49). The aforementioned study generated xenograft tumor metastasis mice and TMEM196 knockout (KO) mice, which strongly supports that TMEM196 can be used as prognostic markers in lung cancer and may represent potential targets for cancer treatment (17). Concurrently, TMEM17 is increased in breast cancer (32) and glioblastoma (33) but decreased in NSCLC (34), suggesting even the same TMEM protein may play different roles in different cancers. With further exploration and research, more different signaling pathways and molecules or unknown mechanisms will be revealed, potentially providing new directions or targets for the treatment of cancers.

3. TMEM and cardiovascular diseases

In recent years, the mechanism of TMEM in cardiovascular diseases has also attracted the attention of numerous researchers, especially its family member TMEM16A, whose role is mainly manifested in pulmonary artery hypertension (PAH) and hypertension (Table II).

Table II

The roles and functions of TMEMs in in various heart diseases.

Table II

The roles and functions of TMEMs in in various heart diseases.

First author/s, yearTMEMDisease typesAssociated tissues or cellsAlterationsPathwayAssociated targetsFunction or mechanism(Refs.)
Liu et al, 2020TMEM16A (ANO1)PAHPulmonary ASMCsUpregulationp38MAPK/ERKPCNA, p38 and ERKPromote cell proliferation(50)
Shang et al, 2020 PAHPulmonary ASMCsUpregulationN/Acyclin E, cyclin D1Accelerate cell cycle(51)
Zheng et al, 2021 Vascular remodelingBasilar ASMCsN/ARhoA/ROCK2 and integrinβ3/FAK RhoA/ROCK2/MLCP/MLC20 and integrinβ3/FAKLimit cells migration, protect vascular remodeling(52)
Lv et al, 2020 Aortic remodelingASMCsDownregulationN/Ap62, Bcl-2, Beclin-1, and VPS34Prevents vascular autophagy and remodeling(53)
Ma et al, 2017 Endothelial dysfunction and hypertensionHUVECs, aortic ECs and ASMCsUpregulationN/ANox2, p22phox, p47phox and p67phoxInduce endothelial dysfunction and hypertension(54)
Li et al, 2020TMEM106BCADHCAECsUpregulationN/AN/AIncrease cell adhesion and migration(56)
Li et al, 2021TMEM98ECs and VSMCs dysfunction diseaseHUVECs and VSMCsUpregulationERK and AKT/GSK3βICAM-1/VCAM-1, cyclin D1 and β-cateninPromote ECs adhesion, VSMCs proliferation and migration(57)
Chen et al, 2023TMEM11Cardiac repair and regenerationCardiomyocytesN/A TMEM11-METTL1-ATF5-INCA1 axisMETTL1, ATF5, INCA1Suppress cardiomyocyte proliferation(58)

[i] TMEM, transmembrane; PAH, pulmonary artery hypertension; CAD, coronary artery disease; VECs, vascular endothelial cells; VSMCs, vascular smooth muscle cells; ASMCs. artery smooth muscle cells; HCAECs, human coronary artery endothelial cells; HUVECs, human umbilical vein endothelial cells; N/A, not available.

TMEM16A and cardiovascular diseases

There is accumulating evidence suggesting that TMEM16A could be a positive regulator of disease progression in PAH. TMEM16A is highly expressed in pulmonary artery smooth muscle cells (PASMCs) of PAH with high pulmonary blood flow, which may activate PASMC and accelerate phosphorylation of EKR and AKT (50), or enhance the expression of cyclin D1 and cyclin E in PASMCs to facilitate the transition from of cell cycle from G1 to S phase (51), participating in pulmonary vascular remodeling and leading to PAH eventually.

Unlike PAH, TMEM16A exert a dual role in hypertension. Vascular smooth muscle cells (VSMCs)-specific TMEM16A transgenic (Tg) mice were generated, prevented from being affected by other cells in systemic TMEM16A Tg mice (52). It was demonstrated that TMEM16A may inhibit the RhoA/ROCK2/MLCP/MLC20 and integrinβ3/FAK signaling pathways via suppressing WNK1, so as to limit the migration of Ang II-induced basilar artery smooth muscle cells, thereby protecting cerebral vascular remodeling in Ang II-induced hypertension in vivo and in vitro studies (52). TMEM16A has the same effect in the aorta of hypertension mice. TMEM16A would downregulate the autophagy degree of VSMCs via regulating the four-way interaction, which inhibits p62/Bcl-2 binding and the formation of Beclin-1/VPS34 complex, thereby coordinately preventing vascular autophagy, inhibiting vascular remodeling and ultimately alleviating elevated blood pressure; this finding is also supported by a previous study using VSMCs-specific TMEM16A Tg mice (53). Conversely, TMEM16A can also promote the activation of NOX2 and add the production of ROS in vascular endothelial cells (ECs), leading to endothelial dysfunction and potentially hypertension. In particular, vascular endothelial-specific TMEM16A KO and-specific TMEM16A Tg mice were generated, respectively from in vivo and in vitro studies, as well as upregulation and downregulation of TMEM16A, comprehensively investigating the specific involvement of TMEM16A in regulating endothelial function, blood pressure and the underlying mechanism (54). Modification of TMEM16A may be a novel therapeutic strategy for endothelial dysfunction-associated diseases (54). Inhibition of the function of TMEM16A with specific TMEM16A inhibitor TMinh-23 can attenuate the vasoconstriction effect in vitro and reduce blood pressure in spontaneously hypertensive rats in vivo (55). Taken together, TMEM16A plays a dual role in some vascular-related diseases and potentially serves as a novel therapeutic target for hypertension, endothelial dysfunction and VSMCs' migration-related cardiovascular diseases such as vascular remodeling.

Other TMEM protein family members and cardiovascular diseases

ANRIL and TMEM106B are both coronary heart disease susceptibility genes, located on chromosome 9p21.3 and 7q21.11 respectively. Elevated expression of ANRIL appear to reduce the adhesion of monocytes to ECs and the trans-endothelial migration ability of monocytes, which plays an important role in limiting the progression of coronary artery disease (CAD), while high expression of TMEM106B can completely offset this effect of ANRIL (56). On the contrary, silencing of TMEM106B can significantly reduce the effect of promoting CAD caused by low expression of ANRIL. TMEM98 encodes a secreted protein that can be expressed and secreted with the induction of platelet-derived growth factor-BB. It can affect the adhesion between monocytes and ECs through regulating the expression of ICAM-1 and VCAM-1. Furthermore, TMEM98 can also trigger the proliferation and migration of VSMCs by influencing the expression of cyclin D1 and β-catenin and regulating the AKT/GSK-3β/Cyclin D1 signaling pathway (57). TMEM11 is located mainly in the mitochondria of heart muscle cells and directly interacts with METTL1 and enhances m(7)G methylation of ATF5 mRNA, thereby increasing ATF5 and INCA1 expression, which is involved in the regulation of cardiomyocyte proliferation via mediating TMEM11-METTL1-ATF5-INCA1 axis. This finding reveals that targeting this axis may serve as a novel therapeutic strategy for promoting cardiac repair and regeneration, supported by TMEM11 KO mice and TMEM11 Tg mice (58).

4. TMEM and kidney diseases

Although the aforementioned sections extensively discuss the relationship between the TMEM proteins family and both tumor and cardiovascular diseases, increasing attention from researchers has been directed towards understanding the association between TMEM and renal disorders. Numerous studies have been conducted to explore this domain (Table III).

Table III

The roles and functions of TMEMs in in various kidney diseases.

Table III

The roles and functions of TMEMs in in various kidney diseases.

First author/s, yearTMEMDisease typesAssociated tissues or cellsAlterationsAssociated targetsFunction or mechanism(Refs.)
Ding et al, 2023TMEM72ccRCC, CKD293T, HeLaDownregulationCOPIIInvolved in protein synthesis(59)
Sachiko et al, 2009TMEM22 (SLC35G2)Renal cell carcinomaRCC13, RCC54UpregulationRas-related protein Rab-37Promote cancer cell growth(60)
Thibodeau et al, 2016; Jiang et al, 2021; Schmit et al, 2019TMEM45AccRCCACHN, RCC4UpregulationN/APromote the proliferation and migration and inhibit the apoptosis(61-63)
Wang et al, 2010; Zhang et al, 2014TMEM174Renal development and carcinomas293TUpregulationAP-1, ERKPromote cell proliferation(64,65)
Miyazaki-Anzai et al, 2022 CKDPCS-400-010DownregulationNPT2ARegulate the homeostasis of phosphorus(80)
Wu et al, 2017TMEM106ARenal cancera786-O, 769-P, ACHN, A-498, A-794DownregulationJNK, caspase3Attenuate cell proliferation, migration and enhance apoptosis(66)
Du et al, 2013TMEM67 (Meckelin)PKD293TN/AJNK, ERK, 4E-BP1Aberrant TMEM67 expression results in abnormal cell proliferation and cyst formation(72)
Zhu et al, 2021 PKDN/ADownregulationmTORTMEM67 mutants aggravate renal cysts(73)
Orphal et al, 2020TMEM63CEMT associated diseases293TDownregulationTGF-β, α-SMA and E-cadherinMaintain cell viability(77)
Schulz et al, 2019 FSGSPodocyteDownregulationNephrinMaintain cell viability and inhibit apoptosis(76)
Faria et al, 2014TMEM16AProximal tubular dysfunctionPTEDownregulationN/AReduce urinary protein excretion(79)
Liu et al, 2021TMEM30AMN, MCD and FSGSPodocyteDownregulationCHOP, PDIMaintain podocyte survival and integrity of GFB(81)

[i] TMEM106:

[ii] aPrimary Renal Proximal Tubule Epithelial Cells, Primary Renal Cortical Epithelial Cells and renal cancer cells (786-O,769-P, ACHN, A-498, A-794). TMEM, transmembrane; CKD, chronic kidney disease; ccRCC, clear cell renal cell carcinoma; FSGS, focal segmental glomerulosclerosis; MN, membranous nephropathy; MCD, minimal change disease; PKD, polycystic kidney disease; EMT, epithelial-mesenchymal transition; α-SMA, α-smooth muscle actin; N/A, not available.

TMEM and clear cell renal cell cancer

In clear cell renal cell carcinoma (ccRCC), TMEM22 and TMEM45A are found to be upregulated, whereas TMEM7, TMEM30B, TMEM45B, TMEM52B, TMEM61, TMEM72, TMEM116, TMEM207 and TMEM213 demonstrate a downregulated state (6). TMEM45A is the most upregulated and TMEM213 is the most downregulated among them. The TMEM72 protein has four transmembrane domains and a long c-terminal tail, located on the plasma membrane. It exerts a vital role in protein folding, assembly and transport in normal kidney development and the pathogenesis of membrane transport-related diseases, especially renal cancer (59). There exists an interaction between TMEM22 and Ras-related protein RAB-37, which highly expresses in renal cell carcinoma. Specific knockdown of TMEM22 by siRNA or downregulation of RAB-37 would significantly slow down the proliferation of cancer cells (60). Therefore, TMEM22 and TMEM72 may be involved in the development, progression and metastasis of renal cell carcinoma.

TMEM45A is upregulated in ccRCC, especially in high grade ccRCC (61), which may be involved in post-translational protein modification and transport. It can promote tumor cell proliferation, EMT and inflammatory response (62) and increase the resistance of RCC4 + pVHL cells to cisplatin by activating the UPR pathway (63). TMEM174 consists of 243 amino acids and contains two transmembrane helices, which determines its subcellular localization in the endoplasmic reticulum and affects its function. It is highly expressed in human normal renal tissues and occassioanlly in patients with renal cancer. Overexpression of TMEM174 can enhance the transcriptional activity of activator protein-1 and promote cell proliferation through the ERK pathway, thus TMEM174 has a potential influence in the normal physiological function and development of kidney and even the occurrence and development of renal carcinoma (64,65). TMEM106A serves as a potential tumor suppressor and is downregulated in renal cancer cells. Its inactivation accelerates proliferation and migration of tumor cells while inhibiting apoptosis (66).

TMEM and polycystic kidney disease

The expression product of TMEM67 is Meckelin protein, which is located on the cell membrane and primary cilia of renal proximal tubular epithelial cells (PTE), which is of great significance for their formation. The normal morphology and function of the cilia are particularly important for the physiological renal function (67,68). Missense mutations of TMEM67 have been detected in patients with cystic kidney disease, indicating that the potential role of Meckelin protein and primary cilia in the development of polycystic kidney disease (PKD) (69-71). Compared with control mice, accumulation of p-ERK, p-JNK, p-4E-BP1 and high level of overall phosphorylation (4G10) are observed in B6C3Fe a/a-bpck mice (with TMEM67 mutation, which displays a severe cystic kidney phenotype and enlarged kidney after birth quickly) in different weeks of age, consistent with the observations of 293T cells in vitro. This finding supports that TMEM67 also mediates PKD through the JNK/ERK signaling pathway (72). However, another group generates TMEM67 mutant zebrafish, which is the first adult zebrafish model of PKD (TMEM67 transcript levels are reduced). The present study has demonstrated that TMEM67 mutants have impaired ciliary function, resulting in abnormal cell structure and signaling, ultimately triggering kidney cysts. Furthermore, there is a hyperactive mTOR signal in the model, and inhibition of this pathway is beneficial to ameliorate PKD in both the embryonic and adult zebrafish models (73). In addition, TMEM107 also plays an important role in the formation and function of cilia, and may be related to PKD, though the underlying mechanisms involved are not yet understood (71,74).

TMEM and other kidney-related diseases

TMEM63 proteins include TMEM63A, TMEM63B and TMEM63C, which together constitute a hyperosmotic activated ion channel and may participate in the osmotic regulation of renal cells and even the whole body (75). TMEM63C may be associated with normal renal function and renal injury in rats and zebrafish (76). MicroRNA (miR)-564 can reduce the expression of TMEM63C in 293T cells and human podocytes (hPC), while TGF-β could add the expression of TMEM63C in a concentration-dependent manner in renal cells (77). In 293T cells, the downregulation of TMEM63C is related to the increase of a-SMA and E-cadherin ratio, which leads to the decreased cell viability (77). In hPC, Ang II may induce the expression of TMEM63C possibly through NF-κB-dependent mechanisms, and TMEM63C is related to the development of proteinuria (78). The decrease of TMEM63C is related to the lowered expression of nephrin, and its inhibition will also lead to impaired cell viability. For example, the downregulation of TMEM63C in patients' podocytes with focal segmental glomerulosclerosis (FSGS) (76). This finding indicates that TMEM63C is a potential hPC survival factor and contributed to maintaining the function of glomerular filtration barrier (77).

TMEM16A is dominantly expressed in human and mouse PTE, with minor expression in podocytes and other tubular segments. In a previous study, three gene models of TMEM16A were generated: systemic KO, podocyte-specific KO and PTE-specific KO mice, fully demonstrating that TMEM16A plays a vital role in PTE rather than podocyte (79). It is a calcium-activated chloride channel in PTE that controls H+ transport by the V-ATPase and uptake of albumin, thereby mediating proton secretion and protein reabsorption. TMEM16A KO decreases urine electrolyte concentrations and increases urine protein excretion (79). TMEM174 is a specific gene expressed in PTC and its corresponding protein interacts with NPT2A in both human and mice, which is a major proximal-specific Pi cotransporter. TMEM174 regulates the homeostasis of Pi by reducing phosphorus uptake and maintaining function of NPT2A. TMEM174 KO mice were generated to demonstrate that NPT2A would be reduced when TMEM174 is deficient in vivo, leading to chronic kidney disease (CKD)-dependent complications such as hyperphosphatemia and vascular calcification (80). The level of TMEM30A is decreased in patients with podocytopathy, including minimal change disease and membranous nephropathy. Podocyte-specific TMEM30A KO mice are generated, which shows, podocyte foot process effacement, lack of a slit diaphragm and increases in the GBM, leading to a series of pathological phenotypic changes such as podocytes injury, proteinuria, mesangial cell proliferation and mesangial matrix accumulation, even glomerulosclerosis ultimately. Mechanistically, podocyte-specific TMEM30A KO may cause endoplasmic reticulum stress via interfering with protein modification and programmed cell death (81).

5. Discussion

In the summary of the association between TMEM proteins and other systemic diseases, it is apparent that the current studies are limited and lack systematic exploration. Concurrently, these studies involve a restricted number of TMEM proteins and a narrowly focused range of diseases. Therefore, the present review does not present a comprehensive summary or discussion concerning TMEM proteins and other systemic diseases. Nonetheless, it is warranting of additional attention that genomic mutations of TMEM genes have been identified, involving single nucleotide polymorphism (SNP) locus or allele point mutation. Predominantly, these mutations are associated with neurodegenerative and immune-related disorders.

The risk variations of TMEM106B are related to some neurodegenerative diseases, particularly SNP locus. Dementia risk allele SNP rs1990622-A predisposes to TMEM106B fibril formation in the hippocampus, and affects brain lipid homeostasis, particularly myelin lipids (82). The risk variation of SNP locus rs1990622 can contribute to Alzheimer's disease (AD) risk and is related to the pathological manifestations of AD, but its T allele is only related to the risk of women, whereas not in men (83). However, variant rs1990621 of TMEM106B may have a neuronal protection effect against general aging, independent of disease status (84). At the same time, the extensive accumulation of C-terminal immunoreactive material of TMEM106B in cases of frontotemporal lobar degeneration (FTLD) with TMEM106B progranulin gene mutations (85). These findings support the pathogenic effect of TMEM106B in a diverse range of neurodegenerative disorders. TMEM gene family is also related to Parkinson's disease (PD). A total of three rare Dmis variants of TMEM230, rare missense variants of TMEM59 in sporadic early onset PD or familial PD. Rare missense variants of TMEM108 appear in sporadic late-onset PD (86).

A total of three SNPs (the CT and CT + TT genotypes in rs12493175, the AC and AC + AA genotypes in rs13062955 and CGTA haplotype) are located in the TMEM39A gene, which is observed to significantly reduce the risk of systemic lupus erythematosus (SLE) in a Chinese Han population (87). A genome-wide association study of SLE susceptibility in European Americans showed that the TMEM39A SNP locus rs1132200 is a susceptibility gene for SLE (88). The major allele G of TMEM187 (rs13397) could be considered as a risk genetic allele for rheumatoid arthritis in Egyptian populations (89). These studies underscore the necessity for expanded investigation into the multifaceted roles of the TMEM gene family across diverse pathological processes.

Some aforementioned studies were supported by genetic models in vivo: TMEM196 in lung cancer (17); TMEM11 in cardiac repair and regeneration (58), VSMC-TMEM16A in hypertension (52-54); PTE-TMEM16A in normal renal function (79), TMEM174 in Pi homeostasis (80) and podocyte-specific TMEM30A in podocytopathy (81). Among them, the results about TMEM proteins as therapeutic targets are limited. For example, targeting TMEM proteins themselves such as TMEM16A (55) and TMEM63C (77), or upstream/downstream molecules dependent on TMEM proteins, such as TMEM63A (90), TMEM229A (19), TMEM17(32) and TMEM11(58).

Inhibition of the function of TMEM16A with specific TMEM16A inhibitor TMinh-23 can attenuate the vasoconstriction effect in vitro and in vivo (55). MiR-564 can reduce the expression of TMEM63C in hPC and 293T cells, thereby decreasing cell viability and increasing EMT in renal cells (77). TMEM63A is localized in endoplasmic reticulum and lysosomal membrane, and interacts with valosin-containing protein (VCP) and its cofactor derlin 1 (DERL1). Pharmacological inhibition of VCP by CB-5083 or knockdown of DERL1 partially eliminated the carcinogenic effect of TMEM63A on triple-negative breast cancer (TNBC) progression in vivo and in vitro (90). TMEM229A is decreased in NSCLC and associated with a poor prognosis in patients. Overexpression of TMEM229A reduces the expression levels of p-ERK and p-AKT, which can be partially suppressed by the specific ERK inhibitor PD98059, suggesting that TMEM229A plays a favorable role in NSCLC via the downstream molecule ERK (19). TMEM17 can promote malignant progression of breast cancer cells by activating the AKT/GSK3β signaling pathway, and these effects reversed by AKT inhibitor LY294002. However, how TMEM17 affects p-AKT remains unclear, and the detailed mechanism should be elucidated in future studies (32). It is TMEM11-METTL1-ATF5-INCA1 axis in cardiomyocyte proliferation that reveals that targeting this axis may serve as a novel therapeutic strategy for promoting cardiac repair and regeneration (58).

In the present review, it was also discussed that some TMEM family members are involved in the occurrence and development of different diseases in multiple systems, such as TMEM63, TMEM98 and TMEM106. TMEM63 includes TMEM63A, TMEM63B and TMEM63C. TMEM63 localizes to lysosomes, mediate lysosomal mechano-sensitivity and modulate lysosomal morphology and function in drosophilae (91). TMEM63A promotes TNBC progression (90). TMEM63C maintains viability of 293T and podocyte and inhibits their apoptosis in EMT-associated diseases and FSGS (76-78). TMEM98 confers chemoresistance of HCC (18) and promotes adhesion of ECs and proliferation and migration of VSMCs in ECs and VSMCs dysfunction related-diseases (57). TMEM106 includes TMEM106A, TMEM106B and TMEM106C. Inactivation of TMEM106A accelerates proliferation and migration of tumor cells while inhibiting apoptosis in renal cancer cells (66). TMEM106B plays an unfavorable role in CAD by mediating another CAD susceptibility gene (56) and risk variations of SNP locus of TMEM106B are related to some neurodegenerative diseases such as AD (83) and FTLD (85). TMEM106C contributes to malignant characteristics and poor prognosis in HCC (92).

In conclusion, members of the TMEM family are diversely distributed across various systems and tissues. The localization and function of proteins encoded by them are also different considerably. They are involved in the normal physiological function of human and animal, and modulate disease onset or progression. For example, they exert their functions in promoting tumor, inhibiting tumor and heterogeneity in the development of neoplasms through different signaling pathways: TMEM17, TMEM45A, TMEM45B, TMEM88, TMEM158 and TMEM97 can mediate the proliferation, adhesion and invasion of various tumors, even affect the chemosensitivity. The TMEM protein family is also involved in the pathogenesis and progression of cardiovascular diseases.

Furthermore, the relationship between TMEM and renal disease has sparked great interest. There are TMEM family members related to the most common ccRCC in renal cancer, which need more comprehensive elaboration. However, the exploration of the relationship between TMEM and CKD such as diabetic nephropathy (DN) remains in its infancy, with few current studies and results available. TMEM256, a number of TMEM families, encodes a protein of ~11.7 kDa (https://www.genecards.org/cgi-bin/carddisp.pl?gene=TMEM256&keywords=TMEM256). TMEM256 is expressed in proximal renal tubular cells, and its subcellular localization is in exosomes. Nonetheless, its underlying structure and function remain uncertain. The role of TMEM256 in DN and how it mediates disease progression have not been studied and further studies are needed.

At present, results about TMEM proteins as therapeutic targets, even researches of drug and rescue, are still lacking either in vivo or in vitro. Especially the efficiency and results of clinical transformation are still few. Very few studies have been reported. For TMEM proteins, the following parameters should be investigated: the structural characteristics of transmembrane proteins, whether they have special roles in hydrophilic, hydrophobic or transmembrane regions, or whether there are specific post-translational modifications on certain amino acid sequences, which are closely related to their functions in the organism. In the future, genetic model animals should be added to studies that have been reported only in vitro to validate the accuracy of results in vitro, involving either Tg or KO animal, either systemic or even specific animal. Signaling pathways or downstream molecules being involved in the occurrence and development of diseases should be confirmed by transcriptomics, proteomics or other predictive methods and experimental verification. Only through such similar processes can corresponding purified proteins, recombinant proteins or agonist/inhibitors be designed to conduct drug intervention or rescue studies, in order to increase the efficiency of clinical transformation and make clinical practice become possible in the future.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the National Natural Science Foundation of China (grant nos. 81873616 and 82170730), the GDPH Supporting Fund for NSFC Program (grant nos. 81873616 and 82170730) the High-level Hospital Construction Project (grant no. KJ012021013), the Guangdong-Hong Kong-Macao-Joint Labs Program from Guangdong Science and Technology (grant no. 2019B121205005) and the 'Group-type' Special Support Project for Education Talents in Universities (grant no. 4SG21227G).

Availability of data and materials

Not applicable.

Authors' contributions

HX designed and wrote the major part of original manuscript. SY, PL, YZ and TZ prepared the tables and revised the manuscript. JLa, HJ, DW, JLi and XB participated in revising and reviewing the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ryu H, Fuwad A, Yoon S, Jang H, Lee JC, Kim SM and Jeon TJ: Biomimetic membranes with transmembrane proteins: State-of-the-art in transmembrane protein applications. Int J Mol Sci. 20(1437)2019.PubMed/NCBI View Article : Google Scholar

2 

Stillwell W: Chapter 6-membrane proteins. In: An introduction to biological membranes (second edition). Stillwell W (ed). Elsevier, pp89-110, 2016.

3 

Marx S, Dal Maso T, Chen JW, Bury M, Wouters J, Michiels C and Le Calvé B: Transmembrane (TMEM) protein family members: Poorly characterized even if essential for the metastatic process. Semin Cancer Biol. 60:96–106. 2020.PubMed/NCBI View Article : Google Scholar

4 

Xu D, Qu L, Hu J, Li G, Lv P, Ma D, Guo M and Chen Y: Transmembrane protein 106A is silenced by promoter region hypermethylation and suppresses gastric cancer growth by inducing apoptosis. J Cell Mol Med. 18:1655–1666. 2014.PubMed/NCBI View Article : Google Scholar

5 

Segelcke D, Fischer HK, Hütte M, Dennerlein S, Benseler F, Brose N, Pogatzki-Zahn EM and Schmidt M: Tmem160 contributes to the establishment of discrete nerve injury-induced pain behaviors in male mice. Cell Rep. 37(110152)2021.PubMed/NCBI View Article : Google Scholar

6 

Wrzesiński T, Szelag M, Cieślikowski WA, Ida A, Giles R, Zodro E, Szumska J, Poźniak J, Kwias Z, Bluyssen HA and Wesoly J: Expression of pre-selected TMEMs with predicted ER localization as potential classifiers of ccRCC tumors. BMC Cancer. 15(518)2015.PubMed/NCBI View Article : Google Scholar

7 

Zhang Z, Luo S, Barbosa GO, Bai M, Kornberg TB and Ma DK: The conserved transmembrane protein TMEM-39 coordinates with COPII to promote collagen secretion and regulate ER stress response. PLoS Genet. 17(e1009317)2021.PubMed/NCBI View Article : Google Scholar

8 

Foulquier F, Amyere M, Jaeken J, Zeevaert R, Schollen E, Race V, Bammens R, Morelle W, Rosnoblet C, Legrand D, et al: TMEM165 deficiency causes a congenital disorder of glycosylation. Am J Hum Genet. 91:15–26. 2012.PubMed/NCBI View Article : Google Scholar

9 

Jung YS, Jun S, Kim MJ, Lee SH, Suh HN, Lien EM, Jung HY, Lee S, Zhang J, Yang JI, et al: TMEM9 promotes intestinal tumorigenesis through vacuolar-ATPase-activated Wnt/β-catenin signalling. Nat Cell Biol. 20:1421–1433. 2018.PubMed/NCBI View Article : Google Scholar

10 

Tropea TF, Mak J, Guo MH, Xie SX, Suh E, Rick J, Siderowf A, Weintraub D, Grossman M, Irwin D, et al: TMEM106B effect on cognition in Parkinson disease and frontotemporal dementia. Ann Neurol. 85:801–811. 2019.PubMed/NCBI View Article : Google Scholar

11 

Wang P, Zhao W, Sun J, Tao T, Chen X, Zheng YY, Zhang CH, Chen Z, Gao YQ, She F, et al: Inflammatory mediators mediate airway smooth muscle contraction through a G protein-coupled receptor-transmembrane protein 16A-voltage-dependent Ca2+ channel axis and contribute to bronchial hyperresponsiveness in asthma. J Allergy Clin Immunol. 141:1259–1268.e11. 2018.PubMed/NCBI View Article : Google Scholar

12 

van der Mark VA, Ghiboub M, Marsman C, Zhao J, van Dijk R, Hiralall JK, Ho-Mok KS, Castricum Z, de Jonge WJ, Oude Elferink RP and Paulusma CC: Phospholipid flippases attenuate LPS-induced TLR4 signaling by mediating endocytic retrieval of Toll-like receptor 4. Cell Mol Life Sci. 74:715–730. 2017.PubMed/NCBI View Article : Google Scholar

13 

Lemmon MA and Schlessinger J: Cell signaling by receptor tyrosine kinases. Cell. 141:1117–1134. 2010.PubMed/NCBI View Article : Google Scholar

14 

Yang J, Chen J, Del Carmen Vitery M, Osei-Owusu J, Chu J, Yu H, Sun S and Qiu Z: PAC, an evolutionarily conserved membrane protein, is a proton-activated chloride channel. Science. 364:395–399. 2019.PubMed/NCBI View Article : Google Scholar

15 

Fu Q, Wu X, Lu Z, Chang Y, Jin Q, Jin T and Zhang M: TMEM205 induces TAM/M2 polarization to promote cisplatin resistance in gastric cancer. Gastric Cancer. 27:998–1015. 2024.PubMed/NCBI View Article : Google Scholar

16 

Guo Q, Shen S, Liao M, Lian P and Wang X: NGX6 inhibits cell invasion and adhesion through suppression of Wnt/beta-catenin signal pathway in colon cancer. Acta Biochim Biophys Sin (Shanghai). 42:450–456. 2010.PubMed/NCBI View Article : Google Scholar

17 

Chen J, Wang D, Chen H, Gu J, Jiang X, Han F, Cao J, Liu W and Liu J: TMEM196 inhibits lung cancer metastasis by regulating the Wnt/β-catenin signaling pathway. J Cancer Res Clin Oncol. 149:653–667. 2023.PubMed/NCBI View Article : Google Scholar

18 

Ng KT, Lo CM, Guo DY, Qi X, Li CX, Geng W, Liu XB, Ling CC, Ma YY, Yeung WH, et al: Identification of transmembrane protein 98 as a novel chemoresistance-conferring gene in hepatocellular carcinoma. Mol Cancer Ther. 13:1285–1297. 2014.PubMed/NCBI View Article : Google Scholar

19 

Zhang X, He Y, Jiang Y, Bao Y, Chen Q, Xie D, Yu H and Wang X: TMEM229A suppresses non-small cell lung cancer progression via inactivating the ERK pathway. Oncol Rep. 46(176)2021.PubMed/NCBI View Article : Google Scholar

20 

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.PubMed/NCBI View Article : Google Scholar

21 

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(e115443)2014.PubMed/NCBI View Article : Google Scholar

22 

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 View Article : Google Scholar

23 

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.PubMed/NCBI View Article : Google Scholar

24 

Duvvuri U, Shiwarski DJ, Xiao D, Bertrand C, Huang X, Edinger RS, Rock JR, Harfe BD, Henson BJ, Kunzelmann K, et al: TMEM16A induces MAPK and contributes directly to tumorigenesis and cancer progression. Cancer Res. 72:3270–3281. 2012.PubMed/NCBI View Article : Google Scholar

25 

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.PubMed/NCBI View Article : Google Scholar

26 

Liu F, Cao QH, Lu DJ, Luo B, Lu XF, Luo RC and Wang XG: TMEM16A overexpression contributes to tumor invasion and poor prognosis of human gastric cancer through TGF-β signaling. Oncotarget. 6:11585–11599. 2015.PubMed/NCBI View Article : Google Scholar

27 

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 Rep. 9:1068–1074. 2014.PubMed/NCBI View Article : Google Scholar

28 

Jia L, Liu W, Guan L, Lu M and Wang K: Inhibition of calcium-activated chloride channel ANO1/TMEM16A suppresses tumor growth and invasion in human lung cancer. PLoS One. 10(e0136584)2015.PubMed/NCBI View Article : Google Scholar

29 

Mazzone A, Eisenman ST, Strege PR, Yao Z, Ordog T, Gibbons SJ and Farrugia G: Inhibition of cell proliferation by a selective inhibitor of the Ca(2+)-activated Cl(-) channel, Ano1. Biochem Biophys Res Commun. 427:248–253. 2012.PubMed/NCBI View Article : Google Scholar

30 

Cha JY, Wee J, Jung J, Jang Y, Lee B, Hong GS, Chang BC, Choi YL, Shin YK, Min HY, et al: Anoctamin 1 (TMEM16A) is essential for testosterone-induced prostate hyperplasia. Proc Natl Acad Sci USA. 112:9722–9727. 2015.PubMed/NCBI View Article : Google Scholar

31 

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.PubMed/NCBI View Article : Google Scholar

32 

Zhao Y, Song K, Zhang Y, Xu H, Zhang X, Wang L, Fan C, Jiang G and Wang E: TMEM17 promotes malignant progression of breast cancer via AKT/GSK3β signaling. Cancer Manag Res. 10:2419–2428. 2018.PubMed/NCBI View Article : Google Scholar

33 

Wang S, Zhou Q, Yan S, Liu C, Li F, Feng D and He M: TMEM17 promotes tumor progression in glioblastoma by activating the PI3K/AKT pathway. Front Biosci (Landmark Ed). 29(285)2024.PubMed/NCBI View Article : Google Scholar

34 

Zhang X, Zhang Y, Miao Y, Zhou H, Jiang G and Wang E: TMEM17 depresses invasion and metastasis in lung cancer cells via ERK signaling pathway. Oncotarget. 8:70685–70694. 2017.PubMed/NCBI View Article : Google Scholar

35 

Flamant L, Roegiers E, Pierre M, Hayez A, Sterpin C, De Backer O, Arnould T, Poumay Y and Michiels C: TMEM45A is essential for hypoxia-induced chemoresistance in breast and liver cancer cells. BMC Cancer. 12(391)2012.PubMed/NCBI View Article : Google Scholar

36 

Guo J, Chen L, Luo N, Yang W, Qu X and Cheng Z: Inhibition of TMEM45A suppresses proliferation, induces cell cycle arrest and reduces cell invasion in human ovarian cancer cells. Oncol Rep. 33:3124–3130. 2015.PubMed/NCBI View Article : Google Scholar

37 

Shen K, Yu W, Yu Y, Liu X and Cui X: Knockdown of TMEM45B inhibits cell proliferation and invasion in gastric cancer. Biomed Pharmacother. 104:576–581. 2018.PubMed/NCBI View Article : Google Scholar

38 

Li Y, Guo W, Liu S, Zhang B, Yu BB, Yang B, Kan SL and Feng SQ: Silencing transmembrane protein 45B (TNEM45B) inhibits proliferation, invasion, and tumorigenesis in osteosarcoma cells. Oncol Res. 25:1021–1026. 2017.PubMed/NCBI View Article : Google Scholar

39 

Zhang X, Yu X, Jiang G, Miao Y, Wang L, Zhang Y, Liu Y, Fan C, Lin X, Dong Q, et al: Cytosolic TMEM88 promotes invasion and metastasis in lung cancer cells by binding DVLS. Cancer Res. 75:4527–4537. 2015.PubMed/NCBI View Article : Google Scholar

40 

Yu X, Zhang X, Zhang Y, Jiang G, Mao X and Jin F: Cytosolic TMEM88 promotes triple-negative breast cancer by interacting with Dvl. Oncotarget. 6:25034–25045. 2015.PubMed/NCBI View Article : Google Scholar

41 

de Leon M, Cardenas H, Vieth E, Emerson R, Segar M, Liu Y, Nephew K and Matei D: Transmembrane protein 88 (TMEM88) promoter hypomethylation is associated with platinum resistance in ovarian cancer. Gynecol Oncol. 142:539–547. 2016.PubMed/NCBI View Article : Google Scholar

42 

Cheng Z, Guo J, Chen L, Luo N, Yang W and Qu X: Overexpression of TMEM158 contributes to ovarian carcinogenesis. J Exp Clin Cancer Res. 34(75)2015.PubMed/NCBI View Article : Google Scholar

43 

Fu Y, Yao N, Ding D, Zhang X, Liu H, Ma L, Shi W, Zhu C and Tang L: TMEM158 promotes pancreatic cancer aggressiveness by activation of TGFβ1 and PI3K/AKT signaling pathway. J Cell Physiol. 235:2761–2775. 2020.PubMed/NCBI View Article : Google Scholar

44 

Xu XY, Zhang LJ, Yu YQ, Zhang XT, Huang WJ, Nie XC and Song GQ: Down-regulated MAC30 expression inhibits proliferation and mobility of human gastric cancer cells. Cell Physiol Biochem. 33:1359–1368. 2014.PubMed/NCBI View Article : Google Scholar

45 

Zhu H, Su Z, Ning J, Zhou L, Tan L, Sayed S, Song J, Wang Z, Li H, Sun Q, et al: Transmembrane protein 97 exhibits oncogenic properties via enhancing LRP6-mediated Wnt signaling in breast cancer. Cell Death Dis. 12(912)2021.PubMed/NCBI View Article : Google Scholar

46 

Mao D, Zhang X, Wang Z, Xu G and Zhang Y: TMEM97 is transcriptionally activated by YY1 and promotes colorectal cancer progression via the GSK-3β/β-catenin signaling pathway. Hum Cell. 35:1535–1546. 2022.PubMed/NCBI View Article : Google Scholar

47 

Xu Y, Tang Y, Xu Q and He W: TMEM97 knockdown inhibits 5-fluorouracil resistance by regulating epithelial-mesenchymal transition and ABC transporter expression via inactivating the Akt/mTOR pathway in 5-fluorouracil-resistant colorectal cancer cells. Chem Biol Drug Des. 103(e14490)2024.PubMed/NCBI View Article : Google Scholar

48 

Liu WB, Han F, Huang YS, Chen HQ, Chen JP, Wang DD, Jiang X, Yin L, Cao J and Liu JY: TMEM196 hypermethylation as a novel diagnostic and prognostic biomarker for lung cancer. Mol Carcinog. 58:474–487. 2019.PubMed/NCBI View Article : Google Scholar

49 

Liu WB, Han F, Jiang X, Chen HQ, Zhao H, Liu Y, Li YH, Huang C, Cao J and Liu JY: TMEM196 acts as a novel functional tumour suppressor inactivated by DNA methylation and is a potential prognostic biomarker in lung cancer. Oncotarget. 6:21225–21239. 2015.PubMed/NCBI View Article : Google Scholar

50 

Liu D, Wang K, Su D, Huang Y, Shang L, Zhao Y, Huang J and Pang Y: TMEM16A regulates pulmonary arterial smooth muscle cells proliferation via p38MAPK/ERK pathway in high pulmonary blood flow-induced pulmonary arterial hypertension. J Vasc Res. 58:27–37. 2020.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print).

51 

Shang L, Wang K, Liu D, Qin S, Huang J, Zhao Y and Pang Y: TMEM16A regulates the cell cycle of pulmonary artery smooth muscle cells in high-flow-induced pulmonary arterial hypertension rat model. Exp Ther Med. 19:3275–3281. 2020.PubMed/NCBI View Article : Google Scholar

52 

Zheng H, Li X, Zeng X, Huang C, Ma M, Lv X, Zhang Y, Sun L, Wang G, Du Y and Guan Y: TMEM16A inhibits angiotensin II-induced basilar artery smooth muscle cell migration in a WNK1-dependent manner. Acta Pharm Sin B. 11:3994–4007. 2021.PubMed/NCBI View Article : Google Scholar

53 

Lv XF, Zhang YJ, Liu X, Zheng HQ, Liu CZ, Zeng XL, Li XY, Lin XC, Lin CX, Ma MM, et al: TMEM16A ameliorates vascular remodeling by suppressing autophagy via inhibiting Bcl-2-p62 complex formation. Theranostics. 10:3980–3993. 2020.PubMed/NCBI View Article : Google Scholar

54 

Ma MM, Gao M, Guo KM, Wang M, Li XY, Zeng XL, Sun L, Lv XF, Du YH, Wang GL, et al: TMEM16A contributes to endothelial dysfunction by facilitating Nox2 NADPH oxidase-derived reactive oxygen species generation in hypertension. Hypertension. 69:892–901. 2017.PubMed/NCBI View Article : Google Scholar

55 

Cil O, Chen X, Askew Page HR, Baldwin SN, Jordan MC, Myat Thwe P, Anderson MO, Haggie PM, Greenwood IA, Roos KP and Verkman AS: A small molecule inhibitor of the chloride channel TMEM16A blocks vascular smooth muscle contraction and lowers blood pressure in spontaneously hypertensive rats. Kidney Int. 100:311–320. 2021.PubMed/NCBI View Article : Google Scholar

56 

Li Y, Cho H, Wang F, Canela-Xandri O, Luo C, Rawlik K, Archacki S, Xu C, Tenesa A, Chen Q and Wang QK: Statistical and functional studies identify epistasis of cardiovascular risk genomic variants from genome-wide association studies. J Am Heart Assoc. 9(e014146)2020.PubMed/NCBI View Article : Google Scholar

57 

Li M, Zhu H, Hu X, Gao F, Hu X, Cui Y, Wei X, Xie C, Lv G, Zhao Y and Gao Y: TMEM98, a novel secretory protein, promotes endothelial cell adhesion as well as vascular smooth muscle cell proliferation and migration. Can J Physiol Pharmacol. 99:536–548. 2021.PubMed/NCBI View Article : Google Scholar

58 

Chen XZ, Li XM, Xu SJ, Hu S, Wang T, Li RF, Liu CY, Xue JQ, Zhou LY, Wang YH, et al: TMEM11 regulates cardiomyocyte proliferation and cardiac repair via METTL1-mediated m7G methylation of ATF5 mRNA. Cell Death Differ. 30:1786–1798. 2023.PubMed/NCBI View Article : Google Scholar

59 

Ding J, Matsumiya T, Miki Y, Hayakari R, Shiba Y, Kawaguchi S, Seya K and Imaizumi T: ER export signals mediate plasma membrane localization of transmembrane protein TMEM72. FEBS J. 290:2636–2657. 2023.PubMed/NCBI View Article : Google Scholar

60 

Dobashi S, Katagiri T, Hirota E, Ashida S, Daigo Y, Shuin T, Fujioka T, Miki T and Nakamura Y: Involvement of TMEM22 overexpression in the growth of renal cell carcinoma cells. Oncol Rep. 21:305–312. 2009.PubMed/NCBI

61 

Thibodeau BJ, Fulton M, Fortier LE, Geddes TJ, Pruetz BL, Ahmed S, Banes-Berceli A, Zhang PL, Wilson GD and Hafron J: Characterization of clear cell renal cell carcinoma by gene expression profiling. Urol Oncol. 34:168.e1–e9. 2016.PubMed/NCBI View Article : Google Scholar

62 

Jiang H, Chen H, Wan P, Liang M and Chen N: Upregulation of TMEM45A promoted the progression of clear cell renal cell carcinoma in vitro. J Inflamm Res. 14:6421–6430. 2021.PubMed/NCBI View Article : Google Scholar

63 

Schmit K, Chen JW, Ayama-Canden S, Fransolet M, Finet L, Demazy C, D'Hondt L, Graux C and Michiels C: Characterization of the role of TMEM45A in cancer cell sensitivity to cisplatin. Cell Death Dis. 10(919)2019.PubMed/NCBI View Article : Google Scholar

64 

Wang P, Sun B, Hao D, Zhang X, Shi T and Ma D: Human TMEM174 that is highly expressed in kidney tissue activates AP-1 and promotes cell proliferation. Biochem Biophys Res Commun. 394:993–999. 2010.PubMed/NCBI View Article : Google Scholar

65 

Zhang X, Hu F, Meng L, Gou L and Luo M: Analysis of TMEM174 gene expression in various renal cancer types by RNA in situ hybridization. Oncol Lett. 8:1693–1696. 2014.PubMed/NCBI View Article : Google Scholar

66 

Wu C, Xu J, Wang H, Zhang J, Zhong J, Zou X, Chen Y, Yang G, Zhong Y, Lai D, et al: TMEM106a is a novel tumor suppressor in human renal cancer. Kidney Blood Press Res. 42:853–864. 2017.PubMed/NCBI View Article : Google Scholar

67 

Dawe HR, Smith UM, Cullinane AR, Gerrelli D, Cox P, Badano JL, Blair-Reid S, Sriram N, Katsanis N, Attie-Bitach T, et al: The meckel-gruber syndrome proteins MKS1 and meckelin interact and are required for primary cilium formation. Hum Mol Genet. 16:173–186. 2007.PubMed/NCBI View Article : Google Scholar

68 

McConnachie DJ, Stow JL and Mallett AJ: Ciliopathies and the kidney: A review. Am J Kidney Dis. 77:410–419. 2021.PubMed/NCBI View Article : Google Scholar

69 

Yoder BK, Hou X and Guay-Woodford LM: The polycystic kidney disease proteins, polycystin-1, polycystin-2, polaris, and cystin, are co-localized in renal cilia. J Am Soc Nephrol. 13:2508–2516. 2002.PubMed/NCBI View Article : Google Scholar

70 

Otto EA, Tory K, Attanasio M, Zhou W, Chaki M, Paruchuri Y, Wise EL, Wolf MT, Utsch B, Becker C, et al: Hypomorphic mutations in meckelin (MKS3/TMEM67) cause nephronophthisis with liver fibrosis (NPHP11). J Med Genet. 46:663–670. 2009.PubMed/NCBI View Article : Google Scholar

71 

Hu HY, Zhang J, Qiu W, Liang C, Li CX, Wei TY, Feng ZK, Guo Q, Yang K and Liu ZG: Comprehensive strategy improves the genetic diagnosis of different polycystic kidney diseases. J Cell Mol Med. 25:6318–6332. 2021.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print).

72 

Du E, Li H, Jin S, Hu X, Qiu M and Han R: Evidence that TMEM67 causes polycystic kidney disease through activation of JNK/ERK-dependent pathways. Cell Biol Int. 37:694–702. 2013.PubMed/NCBI View Article : Google Scholar

73 

Zhu P, Qiu Q, Harris PC, Xu X and Lin X: mtor haploinsufficiency ameliorates renal cysts and cilia abnormality in adult zebrafish tmem67 mutants. J Am Soc Nephrol. 32:822–836. 2021.PubMed/NCBI View Article : Google Scholar

74 

Lambacher NJ, Bruel AL, van Dam TJP, Szymańska K, Slaats GG, Kuhns S, McManus GJ, Kennedy JE, Gaff K, Wu KM, et al: TMEM107 recruits ciliopathy proteins to subdomains of the ciliary transition zone and causes Joubert syndrome. Nat Cell Biol. 18:122–131. 2016.PubMed/NCBI View Article : Google Scholar

75 

Zhao X, Yan X, Liu Y, Zhang P and Ni X: Co-expression of mouse TMEM63A, TMEM63B and TMEM63C confers hyperosmolarity activated ion currents in HEK293 cells. Cell Biochem Funct. 34:238–241. 2016.PubMed/NCBI View Article : Google Scholar

76 

Schulz A, Müller NV, van de Lest NA, Eisenreich A, Schmidbauer M, Barysenka A, Purfürst B, Sporbert A, Lorenzen T, Meyer AM, et al: Analysis of the genomic architecture of a complex trait locus in hypertensive rat models links Tmem63c to kidney damage. Elife. 8(e42068)2019.PubMed/NCBI View Article : Google Scholar

77 

Orphal M, Gillespie A, Böhme K, Subrova J, Eisenreich A and Kreutz R: TMEM63C, a potential novel target for albuminuria development, is regulated by MicroRNA-564 and transforming growth factor beta in human renal cells. Kidney Blood Press Res. 45:850–862. 2020.PubMed/NCBI View Article : Google Scholar

78 

Eisenreich A, Orphal M, Böhme K and Kreutz R: Tmem63c is a potential pro-survival factor in angiotensin II-treated human podocytes. Life Sci. 258(118175)2020.PubMed/NCBI View Article : Google Scholar

79 

Faria D, Rock JR, Romao AM, Schweda F, Bandulik S, Witzgall R, Schlatter E, Heitzmann D, Pavenstädt H, Herrmann E, et al: The calcium-activated chloride channel Anoctamin 1 contributes to the regulation of renal function. Kidney Int. 85:1369–1381. 2014.PubMed/NCBI View Article : Google Scholar

80 

Miyazaki-Anzai S, Keenan AL, Blaine J and Miyazaki M: Targeted disruption of a proximal tubule-specific TMEM174 gene in mice causes hyperphosphatemia and vascular calcification. J Am Soc Nephrol. 33:1477–1486. 2022.PubMed/NCBI View Article : Google Scholar

81 

Liu W, Peng L, Tian W, Li Y, Zhang P, Sun K, Yang Y, Li X, Li G and Zhu X: Loss of phosphatidylserine flippase β-subunit Tmem30a in podocytes leads to albuminuria and glomerulosclerosis. Dis Model Mech. 14(dmm048777)2021.PubMed/NCBI View Article : Google Scholar

82 

Lee JY, Harney DJ, Teo JD, Kwok JB, Sutherland GT, Larance M and Don AS: The major TMEM106B dementia risk allele affects TMEM106B protein levels, fibril formation, and myelin lipid homeostasis in the ageing human hippocampus. Mol Neurodegener. 18(63)2023.PubMed/NCBI View Article : Google Scholar

83 

Hu Y, Sun JY, Zhang Y, Zhang H, Gao S, Wang T, Han Z, Wang L, Sun BL and Liu G: rs1990622 variant associates with Alzheimer's disease and regulates TMEM106B expression in human brain tissues. BMC Med. 19(11)2021.PubMed/NCBI View Article : Google Scholar

84 

Li Z, Farias FHG, Dube U, Del-Aguila JL, Mihindukulasuriya KA, Fernandez MV, Ibanez L, Budde JP, Wang F, Lake AM, et al: The TMEM106B FTLD-protective variant, rs1990621, is also associated with increased neuronal proportion. Acta Neuropathol. 139:45–61. 2020.PubMed/NCBI View Article : Google Scholar

85 

Perneel J, Neumann M, Heeman B, Cheung S, Van den Broeck M, Wynants S, Baker M, Vicente CT, Faura J, Rademakers R and Mackenzie IRA: Accumulation of TMEM106B C-terminal fragments in neurodegenerative disease and aging. Acta Neuropathol. 145:285–302. 2023.PubMed/NCBI View Article : Google Scholar

86 

Zhao Y, Zhang K, Pan H, Wang Y, Zhou X, Xiang Y, Xu Q, Sun Q, Tan J, Yan X, et al: Genetic analysis of six transmembrane protein family genes in Parkinson's disease in a large chinese cohort. Front Aging Neurosci. 14(889057)2022.PubMed/NCBI View Article : Google Scholar

87 

Cai X, Huang W, Liu X, Wang L and Jiang Y: Association of novel polymorphisms in TMEM39A gene with systemic lupus erythematosus in a Chinese Han population. BMC Med Genet. 18(43)2017.PubMed/NCBI View Article : Google Scholar

88 

Lessard CJ, Adrianto I, Ice JA, Wiley GB, Kelly JA, Glenn SB, Adler AJ, Li H, Rasmussen A, Williams AH, et al: Identification of IRF8, TMEM39A, and IKZF3-ZPBP2 as susceptibility loci for systemic lupus erythematosus in a large-scale multiracial replication study. Am J Hum Genet. 90:648–660. 2012.PubMed/NCBI View Article : Google Scholar

89 

Tian J, Sun L, Wan L, Zou H, Chen J and Liu F: TMEM44 as a novel prognostic marker for kidney renal clear cell carcinoma is associated with tumor invasion, migration and immune infiltration. Biochem Genet. 62:1200–1215. 2024.PubMed/NCBI View Article : Google Scholar

90 

Zhang TM, Liao L, Yang SY, Huang MY, Zhang YL, Deng L, Hu SY, Yang F, Zhang FL, Shao ZM and Li DQ: TOLLIP-mediated autophagic degradation pathway links the VCP-TMEM63A-DERL1 signaling axis to triple-negative breast cancer progression. Autophagy. 19:805–821. 2023.PubMed/NCBI View Article : Google Scholar

91 

Li K, Guo Y, Wang Y, Zhu R, Chen W, Cheng T, Zhang X, Jia Y, Liu T, Zhang W, et al: Drosophila TMEM63 and mouse TMEM63A are lysosomal mechanosensory ion channels. Nat Cell Biol. 26:393–403. 2024.PubMed/NCBI View Article : Google Scholar

92 

Duan J, Qian Y, Fu X, Chen M, Liu K, Liu H, Yang J, Liu C and Chang Y: TMEM106C contributes to the malignant characteristics and poor prognosis of hepatocellular carcinoma. Aging (Albany NY). 13:5585–5606. 2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2025
Volume 22 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu H, Yang S, Liu P, Zhang Y, Zhang T, Lan J, Jiang H, Wu D, Li J, Bai X, Bai X, et al: The roles and functions of TMEM protein family members in cancers, cardiovascular and kidney diseases (Review). Biomed Rep 22: 63, 2025.
APA
Xu, H., Yang, S., Liu, P., Zhang, Y., Zhang, T., Lan, J. ... Bai, X. (2025). The roles and functions of TMEM protein family members in cancers, cardiovascular and kidney diseases (Review). Biomedical Reports, 22, 63. https://doi.org/10.3892/br.2025.1941
MLA
Xu, H., Yang, S., Liu, P., Zhang, Y., Zhang, T., Lan, J., Jiang, H., Wu, D., Li, J., Bai, X."The roles and functions of TMEM protein family members in cancers, cardiovascular and kidney diseases (Review)". Biomedical Reports 22.4 (2025): 63.
Chicago
Xu, H., Yang, S., Liu, P., Zhang, Y., Zhang, T., Lan, J., Jiang, H., Wu, D., Li, J., Bai, X."The roles and functions of TMEM protein family members in cancers, cardiovascular and kidney diseases (Review)". Biomedical Reports 22, no. 4 (2025): 63. https://doi.org/10.3892/br.2025.1941