Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1)

Standard

Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1). / Schubert, Desirée; Klein, Marie-Christine; Hassdenteufel, Sarah; Caballero-Oteyza, Andrés; Yang, Linlin; Proietti, Michele; Bulashevska, Alla; Kemming, Janine; Kühn, Johannes; Winzer, Sandra; Rusch, Stephan; Fliegauf, Manfred; Schäffer, Alejandro A; Pfeffer, Stefan; Geiger, Roger; Cavalié, Adolfo; Cao, Hongzhi; Yang, Fang; Li, Yong; Rizzi, Marta; Eibel, Hermann; Kobbe, Robin; Marks, Amy L; Peppers, Brian P; Hostoffer, Robert W; Puck, Jennifer M; Zimmermann, Richard; Grimbacher, Bodo.

In: J ALLERGY CLIN IMMUN, 04.08.2017.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Schubert, D, Klein, M-C, Hassdenteufel, S, Caballero-Oteyza, A, Yang, L, Proietti, M, Bulashevska, A, Kemming, J, Kühn, J, Winzer, S, Rusch, S, Fliegauf, M, Schäffer, AA, Pfeffer, S, Geiger, R, Cavalié, A, Cao, H, Yang, F, Li, Y, Rizzi, M, Eibel, H, Kobbe, R, Marks, AL, Peppers, BP, Hostoffer, RW, Puck, JM, Zimmermann, R & Grimbacher, B 2017, 'Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1)', J ALLERGY CLIN IMMUN. https://doi.org/10.1016/j.jaci.2017.06.042

APA

Schubert, D., Klein, M-C., Hassdenteufel, S., Caballero-Oteyza, A., Yang, L., Proietti, M., Bulashevska, A., Kemming, J., Kühn, J., Winzer, S., Rusch, S., Fliegauf, M., Schäffer, A. A., Pfeffer, S., Geiger, R., Cavalié, A., Cao, H., Yang, F., Li, Y., ... Grimbacher, B. (2017). Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1). J ALLERGY CLIN IMMUN. https://doi.org/10.1016/j.jaci.2017.06.042

Vancouver

Bibtex

@article{ad06a34cf26e4d2bb484b4925e79e06b,
title = "Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1)",
abstract = "BACKGROUND: Primary antibody deficiencies (PADs) are the most frequent primary immunodeficiencies in human subjects. The genetic causes of PADs are largely unknown. Sec61 translocon alpha 1 subunit (SEC61A1) is the major subunit of the Sec61 complex, which is the main polypeptide-conducting channel in the endoplasmic reticulum membrane. SEC61A1 is a target gene of spliced X-box binding protein 1 and strongly induced during plasma cell (PC) differentiation.OBJECTIVE: We identified a novel genetic defect and studied its pathologic mechanism in 11 patients from 2 unrelated families with PADs.METHODS: Whole-exome and targeted sequencing were conducted to identify novel genetic mutations. Functional studies were carried out ex vivo in primary cells of patients and in vitro in different cell lines to assess the effect of SEC61A1 mutations on B-cell differentiation and survival.RESULTS: We investigated 2 families with patients with hypogammaglobulinemia, severe recurrent respiratory tract infections, and normal peripheral B- and T-cell subpopulations. On in vitro stimulation, B cells showed an intrinsic deficiency to develop into PCs. Genetic analysis and targeted sequencing identified novel heterozygous missense (c.254T>A, p.V85D) and nonsense (c.1325G>T, p.E381*) mutations in SEC61A1, segregating with the disease phenotype. SEC61A1-V85D was deficient in cotranslational protein translocation, and it disturbed the cellular calcium homeostasis in HeLa cells. Moreover, SEC61A1-V85D triggered the terminal unfolded protein response in multiple myeloma cell lines.CONCLUSION: We describe a monogenic defect leading to a specific PC deficiency in human subjects, expanding our knowledge about the pathogenesis of antibody deficiencies.",
keywords = "Journal Article",
author = "Desir{\'e}e Schubert and Marie-Christine Klein and Sarah Hassdenteufel and Andr{\'e}s Caballero-Oteyza and Linlin Yang and Michele Proietti and Alla Bulashevska and Janine Kemming and Johannes K{\"u}hn and Sandra Winzer and Stephan Rusch and Manfred Fliegauf and Sch{\"a}ffer, {Alejandro A} and Stefan Pfeffer and Roger Geiger and Adolfo Cavali{\'e} and Hongzhi Cao and Fang Yang and Yong Li and Marta Rizzi and Hermann Eibel and Robin Kobbe and Marks, {Amy L} and Peppers, {Brian P} and Hostoffer, {Robert W} and Puck, {Jennifer M} and Richard Zimmermann and Bodo Grimbacher",
note = "Copyright {\textcopyright} 2017 American Academy of Allergy, Asthma & Immunology. All rights reserved.",
year = "2017",
month = aug,
day = "4",
doi = "10.1016/j.jaci.2017.06.042",
language = "English",
journal = "J ALLERGY CLIN IMMUN",
issn = "0091-6749",
publisher = "Mosby Inc.",

}

RIS

TY - JOUR

T1 - Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1)

AU - Schubert, Desirée

AU - Klein, Marie-Christine

AU - Hassdenteufel, Sarah

AU - Caballero-Oteyza, Andrés

AU - Yang, Linlin

AU - Proietti, Michele

AU - Bulashevska, Alla

AU - Kemming, Janine

AU - Kühn, Johannes

AU - Winzer, Sandra

AU - Rusch, Stephan

AU - Fliegauf, Manfred

AU - Schäffer, Alejandro A

AU - Pfeffer, Stefan

AU - Geiger, Roger

AU - Cavalié, Adolfo

AU - Cao, Hongzhi

AU - Yang, Fang

AU - Li, Yong

AU - Rizzi, Marta

AU - Eibel, Hermann

AU - Kobbe, Robin

AU - Marks, Amy L

AU - Peppers, Brian P

AU - Hostoffer, Robert W

AU - Puck, Jennifer M

AU - Zimmermann, Richard

AU - Grimbacher, Bodo

N1 - Copyright © 2017 American Academy of Allergy, Asthma & Immunology. All rights reserved.

PY - 2017/8/4

Y1 - 2017/8/4

N2 - BACKGROUND: Primary antibody deficiencies (PADs) are the most frequent primary immunodeficiencies in human subjects. The genetic causes of PADs are largely unknown. Sec61 translocon alpha 1 subunit (SEC61A1) is the major subunit of the Sec61 complex, which is the main polypeptide-conducting channel in the endoplasmic reticulum membrane. SEC61A1 is a target gene of spliced X-box binding protein 1 and strongly induced during plasma cell (PC) differentiation.OBJECTIVE: We identified a novel genetic defect and studied its pathologic mechanism in 11 patients from 2 unrelated families with PADs.METHODS: Whole-exome and targeted sequencing were conducted to identify novel genetic mutations. Functional studies were carried out ex vivo in primary cells of patients and in vitro in different cell lines to assess the effect of SEC61A1 mutations on B-cell differentiation and survival.RESULTS: We investigated 2 families with patients with hypogammaglobulinemia, severe recurrent respiratory tract infections, and normal peripheral B- and T-cell subpopulations. On in vitro stimulation, B cells showed an intrinsic deficiency to develop into PCs. Genetic analysis and targeted sequencing identified novel heterozygous missense (c.254T>A, p.V85D) and nonsense (c.1325G>T, p.E381*) mutations in SEC61A1, segregating with the disease phenotype. SEC61A1-V85D was deficient in cotranslational protein translocation, and it disturbed the cellular calcium homeostasis in HeLa cells. Moreover, SEC61A1-V85D triggered the terminal unfolded protein response in multiple myeloma cell lines.CONCLUSION: We describe a monogenic defect leading to a specific PC deficiency in human subjects, expanding our knowledge about the pathogenesis of antibody deficiencies.

AB - BACKGROUND: Primary antibody deficiencies (PADs) are the most frequent primary immunodeficiencies in human subjects. The genetic causes of PADs are largely unknown. Sec61 translocon alpha 1 subunit (SEC61A1) is the major subunit of the Sec61 complex, which is the main polypeptide-conducting channel in the endoplasmic reticulum membrane. SEC61A1 is a target gene of spliced X-box binding protein 1 and strongly induced during plasma cell (PC) differentiation.OBJECTIVE: We identified a novel genetic defect and studied its pathologic mechanism in 11 patients from 2 unrelated families with PADs.METHODS: Whole-exome and targeted sequencing were conducted to identify novel genetic mutations. Functional studies were carried out ex vivo in primary cells of patients and in vitro in different cell lines to assess the effect of SEC61A1 mutations on B-cell differentiation and survival.RESULTS: We investigated 2 families with patients with hypogammaglobulinemia, severe recurrent respiratory tract infections, and normal peripheral B- and T-cell subpopulations. On in vitro stimulation, B cells showed an intrinsic deficiency to develop into PCs. Genetic analysis and targeted sequencing identified novel heterozygous missense (c.254T>A, p.V85D) and nonsense (c.1325G>T, p.E381*) mutations in SEC61A1, segregating with the disease phenotype. SEC61A1-V85D was deficient in cotranslational protein translocation, and it disturbed the cellular calcium homeostasis in HeLa cells. Moreover, SEC61A1-V85D triggered the terminal unfolded protein response in multiple myeloma cell lines.CONCLUSION: We describe a monogenic defect leading to a specific PC deficiency in human subjects, expanding our knowledge about the pathogenesis of antibody deficiencies.

KW - Journal Article

U2 - 10.1016/j.jaci.2017.06.042

DO - 10.1016/j.jaci.2017.06.042

M3 - SCORING: Journal article

C2 - 28782633

JO - J ALLERGY CLIN IMMUN

JF - J ALLERGY CLIN IMMUN

SN - 0091-6749

ER -