Metabolic insights from a GHSR-A203E mutant mouse model

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Metabolic insights from a GHSR-A203E mutant mouse model. / Torz, Lola J.; Osborne-Lawrence, Sherri; Rodriguez, Juan; He, Zhenyan; Paula Cornejo, Maria; Roman Mustafa, Emilio; Jin, Chunyu; Petersen, Natalia; Hedegaard, Morten A.; Nybo, Maja; Martinez Damonte, Valentina; Metzger, Nathan P.; Mani, Bharath K.; Williams, Kevin W.; Raingo, Jesica; Perello, Mario; Holst, Birgitte; Zigman, Jeffrey M.

In: Molecular Metabolism, Vol. 39, 101004, 2020.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Torz, LJ, Osborne-Lawrence, S, Rodriguez, J, He, Z, Paula Cornejo, M, Roman Mustafa, E, Jin, C, Petersen, N, Hedegaard, MA, Nybo, M, Martinez Damonte, V, Metzger, NP, Mani, BK, Williams, KW, Raingo, J, Perello, M, Holst, B & Zigman, JM 2020, 'Metabolic insights from a GHSR-A203E mutant mouse model', Molecular Metabolism, vol. 39, 101004. https://doi.org/10.1016/j.molmet.2020.101004

APA

Torz, L. J., Osborne-Lawrence, S., Rodriguez, J., He, Z., Paula Cornejo, M., Roman Mustafa, E., Jin, C., Petersen, N., Hedegaard, M. A., Nybo, M., Martinez Damonte, V., Metzger, N. P., Mani, B. K., Williams, K. W., Raingo, J., Perello, M., Holst, B., & Zigman, J. M. (2020). Metabolic insights from a GHSR-A203E mutant mouse model. Molecular Metabolism, 39, [101004]. https://doi.org/10.1016/j.molmet.2020.101004

Vancouver

Torz LJ, Osborne-Lawrence S, Rodriguez J, He Z, Paula Cornejo M, Roman Mustafa E et al. Metabolic insights from a GHSR-A203E mutant mouse model. Molecular Metabolism. 2020;39. 101004. https://doi.org/10.1016/j.molmet.2020.101004

Author

Torz, Lola J. ; Osborne-Lawrence, Sherri ; Rodriguez, Juan ; He, Zhenyan ; Paula Cornejo, Maria ; Roman Mustafa, Emilio ; Jin, Chunyu ; Petersen, Natalia ; Hedegaard, Morten A. ; Nybo, Maja ; Martinez Damonte, Valentina ; Metzger, Nathan P. ; Mani, Bharath K. ; Williams, Kevin W. ; Raingo, Jesica ; Perello, Mario ; Holst, Birgitte ; Zigman, Jeffrey M. / Metabolic insights from a GHSR-A203E mutant mouse model. In: Molecular Metabolism. 2020 ; Vol. 39.

Bibtex

@article{8b4dd6acbdab49cf9c39f4e6bb5c0d58,
title = "Metabolic insights from a GHSR-A203E mutant mouse model",
abstract = "Objective: Binding of ghrelin to its receptor, growth hormone secretagogue receptor (GHSR), stimulates GH release, induces eating, and increases blood glucose. These processes may also be influenced by constitutive (ghrelin-independent) GHSR activity, as suggested by findings in short people with naturally occurring GHSR-A204E mutations and reduced food intake and blood glucose in rodents administered GHSR inverse agonists, both of which impair constitutive GHSR activity. In this study, we aimed to more fully determine the physiologic relevance of constitutive GHSR activity.Methods: We generated mice with a GHSR mutation that replaces alanine at position 203 with glutamate (GHSR-A203E), which corresponds to the previously described human GHSR-A204E mutation, and used them to conduct ex vivo neuronal electrophysiology and in vivo metabolic assessments. We also measured signaling within COS-7 and HEK293T cells transfected with wild-type GHSR (GHSR-WT) or GHSR-A203E constructs.Results: In COS-7 cells, GHSR-A203E resulted in lower baseline IP3 accumulation than GHSR-WT; ghrelin-induced IP3 accumulation was observed in both constructs. In HEK293T cells co-transfected with voltage-gated Ca(V)2.2 calcium channel complex, GHSR-A203E had no effect on basal Ca(V)2.2 current density while GHSR-WT did; both GHSR-A203E and GHSR-WT inhibited Ca(V)2.2 current in the presence of ghrelin. In cultured hypothalamic neurons from GHSR-A203E and GHSR-deficient mice, native calcium currents were greater than those in neurons from wild-type mice; ghrelin inhibited calcium currents in cultured hypothalamic neurons from both GHSR-A203E and wild-type mice. In brain slices, resting membrane potentials of arcuate NPY neurons from GHSR-A203E mice were hyperpolarized compared to those from wild-type mice; the same percentage of arcuate NPY neurons from GHSR-A203E and wild-type mice depolarized upon ghrelin exposure. The GHSR-A203E mutation did not significantly affect body weight, body length, or femur length in the firstw6 months of life, yet these parameters were lower in GHSR-A203E mice after 1 year of age. During a 7-d 60% caloric restriction regimen, GHSR-A203E mice lacked the usual marked rise in plasma GH and demonstrated an exaggerated drop in blood glucose. Administered ghrelin also exhibited reduced orexigenic and GH secretagogue efficacies in GHSR-A203E mice.Conclusions: Our data suggest that the A203E mutation ablates constitutive GHSR activity and that constitutive GHSR activity contributes to the native depolarizing conductance of GHSR-expressing arcuate NPY neurons. Although the A203E mutation does not block ghrelin-evoked signaling as assessed using in vitro and ex vivo models, GHSR-A203E mice lack the usual acute food intake response to administered ghrelin in vivo. The GHSR-A203E mutation also blunts GH release, and in aged mice leads to reduced body length and femur length, which are consistent with the short stature of human carriers of the GHSR-A204E mutation. (C) 2020 The Author(s). Published by Elsevier GmbH.",
keywords = "Ghrelin, Constitutive activity, Growth hormone, GH, HORMONE SECRETAGOGUE RECEPTOR, GHRELIN RECEPTOR, CONSTITUTIVE ACTIVITY, CALCIUM-CHANNELS, NEURONS, EXPRESSION, ARCUATE, MICE, IDENTIFICATION, INACTIVATION",
author = "Torz, {Lola J.} and Sherri Osborne-Lawrence and Juan Rodriguez and Zhenyan He and {Paula Cornejo}, Maria and {Roman Mustafa}, Emilio and Chunyu Jin and Natalia Petersen and Hedegaard, {Morten A.} and Maja Nybo and {Martinez Damonte}, Valentina and Metzger, {Nathan P.} and Mani, {Bharath K.} and Williams, {Kevin W.} and Jesica Raingo and Mario Perello and Birgitte Holst and Zigman, {Jeffrey M.}",
year = "2020",
doi = "10.1016/j.molmet.2020.101004",
language = "English",
volume = "39",
journal = "Molecular Metabolism",
issn = "2212-8778",
publisher = "Elsevier",

}

RIS

TY - JOUR

T1 - Metabolic insights from a GHSR-A203E mutant mouse model

AU - Torz, Lola J.

AU - Osborne-Lawrence, Sherri

AU - Rodriguez, Juan

AU - He, Zhenyan

AU - Paula Cornejo, Maria

AU - Roman Mustafa, Emilio

AU - Jin, Chunyu

AU - Petersen, Natalia

AU - Hedegaard, Morten A.

AU - Nybo, Maja

AU - Martinez Damonte, Valentina

AU - Metzger, Nathan P.

AU - Mani, Bharath K.

AU - Williams, Kevin W.

AU - Raingo, Jesica

AU - Perello, Mario

AU - Holst, Birgitte

AU - Zigman, Jeffrey M.

PY - 2020

Y1 - 2020

N2 - Objective: Binding of ghrelin to its receptor, growth hormone secretagogue receptor (GHSR), stimulates GH release, induces eating, and increases blood glucose. These processes may also be influenced by constitutive (ghrelin-independent) GHSR activity, as suggested by findings in short people with naturally occurring GHSR-A204E mutations and reduced food intake and blood glucose in rodents administered GHSR inverse agonists, both of which impair constitutive GHSR activity. In this study, we aimed to more fully determine the physiologic relevance of constitutive GHSR activity.Methods: We generated mice with a GHSR mutation that replaces alanine at position 203 with glutamate (GHSR-A203E), which corresponds to the previously described human GHSR-A204E mutation, and used them to conduct ex vivo neuronal electrophysiology and in vivo metabolic assessments. We also measured signaling within COS-7 and HEK293T cells transfected with wild-type GHSR (GHSR-WT) or GHSR-A203E constructs.Results: In COS-7 cells, GHSR-A203E resulted in lower baseline IP3 accumulation than GHSR-WT; ghrelin-induced IP3 accumulation was observed in both constructs. In HEK293T cells co-transfected with voltage-gated Ca(V)2.2 calcium channel complex, GHSR-A203E had no effect on basal Ca(V)2.2 current density while GHSR-WT did; both GHSR-A203E and GHSR-WT inhibited Ca(V)2.2 current in the presence of ghrelin. In cultured hypothalamic neurons from GHSR-A203E and GHSR-deficient mice, native calcium currents were greater than those in neurons from wild-type mice; ghrelin inhibited calcium currents in cultured hypothalamic neurons from both GHSR-A203E and wild-type mice. In brain slices, resting membrane potentials of arcuate NPY neurons from GHSR-A203E mice were hyperpolarized compared to those from wild-type mice; the same percentage of arcuate NPY neurons from GHSR-A203E and wild-type mice depolarized upon ghrelin exposure. The GHSR-A203E mutation did not significantly affect body weight, body length, or femur length in the firstw6 months of life, yet these parameters were lower in GHSR-A203E mice after 1 year of age. During a 7-d 60% caloric restriction regimen, GHSR-A203E mice lacked the usual marked rise in plasma GH and demonstrated an exaggerated drop in blood glucose. Administered ghrelin also exhibited reduced orexigenic and GH secretagogue efficacies in GHSR-A203E mice.Conclusions: Our data suggest that the A203E mutation ablates constitutive GHSR activity and that constitutive GHSR activity contributes to the native depolarizing conductance of GHSR-expressing arcuate NPY neurons. Although the A203E mutation does not block ghrelin-evoked signaling as assessed using in vitro and ex vivo models, GHSR-A203E mice lack the usual acute food intake response to administered ghrelin in vivo. The GHSR-A203E mutation also blunts GH release, and in aged mice leads to reduced body length and femur length, which are consistent with the short stature of human carriers of the GHSR-A204E mutation. (C) 2020 The Author(s). Published by Elsevier GmbH.

AB - Objective: Binding of ghrelin to its receptor, growth hormone secretagogue receptor (GHSR), stimulates GH release, induces eating, and increases blood glucose. These processes may also be influenced by constitutive (ghrelin-independent) GHSR activity, as suggested by findings in short people with naturally occurring GHSR-A204E mutations and reduced food intake and blood glucose in rodents administered GHSR inverse agonists, both of which impair constitutive GHSR activity. In this study, we aimed to more fully determine the physiologic relevance of constitutive GHSR activity.Methods: We generated mice with a GHSR mutation that replaces alanine at position 203 with glutamate (GHSR-A203E), which corresponds to the previously described human GHSR-A204E mutation, and used them to conduct ex vivo neuronal electrophysiology and in vivo metabolic assessments. We also measured signaling within COS-7 and HEK293T cells transfected with wild-type GHSR (GHSR-WT) or GHSR-A203E constructs.Results: In COS-7 cells, GHSR-A203E resulted in lower baseline IP3 accumulation than GHSR-WT; ghrelin-induced IP3 accumulation was observed in both constructs. In HEK293T cells co-transfected with voltage-gated Ca(V)2.2 calcium channel complex, GHSR-A203E had no effect on basal Ca(V)2.2 current density while GHSR-WT did; both GHSR-A203E and GHSR-WT inhibited Ca(V)2.2 current in the presence of ghrelin. In cultured hypothalamic neurons from GHSR-A203E and GHSR-deficient mice, native calcium currents were greater than those in neurons from wild-type mice; ghrelin inhibited calcium currents in cultured hypothalamic neurons from both GHSR-A203E and wild-type mice. In brain slices, resting membrane potentials of arcuate NPY neurons from GHSR-A203E mice were hyperpolarized compared to those from wild-type mice; the same percentage of arcuate NPY neurons from GHSR-A203E and wild-type mice depolarized upon ghrelin exposure. The GHSR-A203E mutation did not significantly affect body weight, body length, or femur length in the firstw6 months of life, yet these parameters were lower in GHSR-A203E mice after 1 year of age. During a 7-d 60% caloric restriction regimen, GHSR-A203E mice lacked the usual marked rise in plasma GH and demonstrated an exaggerated drop in blood glucose. Administered ghrelin also exhibited reduced orexigenic and GH secretagogue efficacies in GHSR-A203E mice.Conclusions: Our data suggest that the A203E mutation ablates constitutive GHSR activity and that constitutive GHSR activity contributes to the native depolarizing conductance of GHSR-expressing arcuate NPY neurons. Although the A203E mutation does not block ghrelin-evoked signaling as assessed using in vitro and ex vivo models, GHSR-A203E mice lack the usual acute food intake response to administered ghrelin in vivo. The GHSR-A203E mutation also blunts GH release, and in aged mice leads to reduced body length and femur length, which are consistent with the short stature of human carriers of the GHSR-A204E mutation. (C) 2020 The Author(s). Published by Elsevier GmbH.

KW - Ghrelin

KW - Constitutive activity

KW - Growth hormone

KW - GH

KW - HORMONE SECRETAGOGUE RECEPTOR

KW - GHRELIN RECEPTOR

KW - CONSTITUTIVE ACTIVITY

KW - CALCIUM-CHANNELS

KW - NEURONS

KW - EXPRESSION

KW - ARCUATE

KW - MICE

KW - IDENTIFICATION

KW - INACTIVATION

U2 - 10.1016/j.molmet.2020.101004

DO - 10.1016/j.molmet.2020.101004

M3 - Journal article

C2 - 32339772

VL - 39

JO - Molecular Metabolism

JF - Molecular Metabolism

SN - 2212-8778

M1 - 101004

ER -

ID: 248332237