The satiety hormone cholecystokinin gates reproduction in fish by controlling gonadotropin secretion

  1. Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
  2. Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, France
  3. BioCampus Montpellier, University of Montpellier, CNRS, INSERM, F-34094 Montpellier, France
  4. The Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
  5. Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O.B 15159, Rishon Letziyon 7505101, Israel

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a provisional response from the authors.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Gáspár Jékely
    Heidelberg University, Heidelberg, Germany
  • Senior Editor
    Didier Stainier
    Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany

Reviewer #1 (Public Review):

Summary:

The pituitary gonadotropins, FSH and LH, are critical regulators of reproduction. In mammals, synthesis and secretion of FSH and LH by gonadotrope cells are controlled by the hypothalamic peptide, GnRH. As FSH and LH are made in the same cells in mammals, variation in the nature of GnRH secretion is thought to contribute to the differential regulation of the two hormones. In contrast, in fish, FSH and LH are produced in distinct gonadotrope populations and may be less (or differently) dependent on GnRH than in mammals. In the present manuscript, the authors endeavored to determine whether FSH may be independently controlled by a distinct peptide, cholecystokinin (CCK), in zebrafish.

Strengths:

The authors demonstrated that the CCK receptor is enriched in FSH-producing relative to LH-producing gonadotropes, and that genetic deletion of the receptor leads to dramatic decreases in gonadotropin production and gonadal development in zebrafish. Also, using innovative in vivo and ex vivo calcium imaging approaches, they show that LH- and FSH-producing gonadotropes preferentially respond to GnRH and CCK, respectively. Exogenous CCK also preferentially stimulated FSH secretion ex vivo and in vivo.

Weaknesses:

The concept that there may be a distinct FSH-releasing hormone (FSHRH) has been debated for decades. As the authors suggest that CCK is the long-sought FSHRH (at least in fish), they must provide data that convincingly leads to such a conclusion. In my estimation, they have not yet met this burden. In particular, they show that CCK is sufficient to activate FSH-producing cells, but have not yet demonstrated its necessity. Their one attempt to do so was using fish in which they inactivated the CCK receptor using CRISPR-Cas9. While this manipulation led to a reduction in FSH, LH was affected to a similar extent. As a result, they have not shown that CCK is a selective regulator of FSH. Moreover, they do not yet demonstrate that the effects observed reflect the loss of the receptor's function in gonadotropes, as opposed to other cell types. It also is not clear whether the phenotypes of the fish reflect perturbations in pituitary development vs. a loss of CCK receptor function in the pituitary later in life. Ideally, the authors would attempt to block CCK signaling in adult fish that develop normally. For example, if CCK receptor antagonists are available, they could be used to treat fish and see whether and how this affects FSH vs. LH secretion.

In the Discussion, the authors suggest that CCK, as a satiety factor, may provide a link between metabolism and reproduction. This is an interesting idea, but it is not supported by the data presented. That is, none of the results shown link metabolic state to CCK regulation of FSH and fertility. Absent such data, the lengthy discussion of the link is speculative and not fully merited.

Also in the Discussion, the authors argue that "CCK directly controls FSH cells by innervating the pituitary gland and binding to specific receptors that are particularly abundant in FSH gonadotrophs." However, their imaging does not demonstrate innervation of FSH cells by CCK terminals (e.g., at the EM level). Moreover, they have not demonstrated the binding of CCK to these cells. Indeed, no CCK receptor protein data are shown. The calcium responses of FSH cells to exogenous CCK certainly suggest the presence of functional CCK receptors therein; but, the nature of the preparations (with all pituitary cell types present) does not demonstrate that CCK is acting directly in these cells. Indeed, the asynchrony in responses of individual FSH cells to CCK (Figure 4) suggests that not all cells may be activated in the same way. Contrast the response of LH cells to GnRH, where the onset of calcium signaling is similar across cells (Figure 3). Finally, as the authors note in the Discussion, the data presented do not enable them to conclude that the endogenous CCK regulating FSH (assuming it does) is from the brain as opposed to other sources (e.g., the gut).

Reviewer #2 (Public Review):

Summary:

This manuscript builds on previous work suggesting that the CCK peptide is the releasing hormone for FSH in fishes, which is different than that observed in mammals where both LH and FSH release are under the control of GnRH. Based on data using calcium imaging as a readout for stimulation of the gonadotrophs, the researchers present data supporting the hypothesis that CCK stimulates FSH-containing cells in the pituitary. In contrast, LH-containing cells show a weak and variable response to CCK but are highly responsive to GnRH. Data are presented that support the role of CCK in the release of FSH. Researchers also state that functional overlap exists in the potency of GnRH to activate FSH cells, thus the two signalling pathways are not separate.

The results are of interest to the field because for many years the assumption has been that fishes use the same signalling mechanism. These data present an intriguing variation where a hormone involved in satiation acts in the control of reproduction.

Strengths:

The strengths of the manuscript are that researchers have shed light on different pathways controlling reproduction in fishes.

Weaknesses:

Weaknesses are that it is not clear if multiple ligand/receptors are involved (more than one CCK and more than one receptor?). The imaging of the CCK terminals and CCK receptors needs to be reinforced.

Reviewer consultation summary:

- The data presented establish sufficiency, but not necessity of CCK in FSH regulation. The paper did not show that CCK endogenously regulates FSH in fish. This has not been established yet.

- The paper presents the pharmacological effects of CCK on ex vivo preparations but does not establish the in vivo physiological function of the peptide. The current evidence for a novel physiological regulatory mechanism is incomplete and would require further physiological experiments. These could include the use of a CCK receptor antagonist in adult fish to see the effects on FSH and LH release, the generation of a CCK knockout, or cell-specific genetic manipulations.

- Zebrafish have two CCK ligands: ccka, cckb and also multiple receptors: cckar, cckbra and cckbrb. There is ambiguity about which CCK receptor and ligand are expressed and which gene was knocked out.

- Blocking CCK action in fish (with receptor KO) affects FSH and LH. Therefore, the work did not demonstrate a selective role for CCK in FSH regulation in vivo and any claims to have discovered FSHRH need to be more conservative.

- The labelling of the terminals with anti-CCK looks a lot like the background and the authors did not show a specificity control (e.g. anti-CCK antibody pre-absorbed with the peptide or anti-CCK in morphant/KO animals).

Author response:

We would first like to thank the editor for considering our findings for publication in eLife. Furthermore, we thank the reviewers and editors for their encouraging reviews and for providing helpful and insightful comments.

Reviewer #1 (Public Review):

Summary:

The pituitary gonadotropins, FSH and LH, are critical regulators of reproduction. In mammals, synthesis and secretion of FSH and LH by gonadotrope cells are controlled by the hypothalamic peptide, GnRH. As FSH and LH are made in the same cells in mammals, variation in the nature of GnRH secretion is thought to contribute to the differential regulation of the two hormones. In contrast, in fish, FSH and LH are produced in distinct gonadotrope populations and may be less (or differently) dependent on GnRH than in mammals. In the present manuscript, the authors endeavored to determine whether FSH may be independently controlled by a distinct peptide, cholecystokinin (CCK), in zebrafish.

Strengths:

The authors demonstrated that the CCK receptor is enriched in FSH-producing relative to LH-producing gonadotropes, and that genetic deletion of the receptor leads to dramatic decreases in gonadotropin production and gonadal development in zebrafish. Also, using innovative in vivo and ex vivo calcium imaging approaches, they show that LH- and FSH-producing gonadotropes preferentially respond to GnRH and CCK, respectively. Exogenous CCK also preferentially stimulated FSH secretion ex vivo and in vivo.

Weaknesses:

The concept that there may be a distinct FSH-releasing hormone (FSHRH) has been debated for decades. As the authors suggest that CCK is the long-sought FSHRH (at least in fish), they must provide data that convincingly leads to such a conclusion. In my estimation, they have not yet met this burden. In particular, they show that CCK is sufficient to activate FSH-producing cells, but have not yet demonstrated its necessity. Their one attempt to do so was using fish in which they inactivated the CCK receptor using CRISPR-Cas9. While this manipulation led to a reduction in FSH, LH was affected to a similar extent. As a result, they have not shown that CCK is a selective regulator of FSH.

Our conclusion regarding the necessity of CCK signaling for FSH secretion is based on the following evidence:

(1) CCK-like receptors are expressed in the pituitary gland predominantly on FSH cells.

(2) Application of CCK to pituitaries elicits FSH cell activation and FSH release, and, to a lesser degree, activation of LH cells.

(3) Mutating the CCK-like receptor causes a decrease in fsh and lh mRNA synthesis.

(4) Mutating the CCK-like receptor gives rise to a phenotype which is identical to that caused by mutation of both lh and fsh genes in zebrafish.

(5) Mutating the FSH-specific CCK receptor in a different species of fish (medaka) also causes a complete shutdown of FSH production and phenocopies a fsh-mutant phenotype (Uehara et al, BioRxiv, DOI: 10.1101/2023.05.26.542428).

Taken together, we believe that this data strongly supports the conclusion that CCK is necessary for FSH production and release from the fish pituitary. Admittedly, the overlapping effects of CCK on both FSH and LH cells in zebrafish (evident in both our calcium imaging experiments and the KO phenotype) complicates the interpretation of the phenotype. We speculate that the effect of CCK on LH cells in zebrafish can be caused either by paracrine signaling within the gland or by the effects of CCK on higher levels of the axis. In our revised manuscript we will make sure to highlight the overlapping effects of CCK on LH cells rather than portray it as a selective activator of FSH cells.

Moreover, they do not yet demonstrate that the effects observed reflect the loss of the receptor's function in gonadotropes, as opposed to other cell types.

Although there is evidence for the expression of CCK receptor in other tissues, we do show a direct decrease of FSH and LH expression in the gonadotrophs of the pituitary of the mutant fish; taken together with its significant expression in FSH cells, it is the most reasonable and forward explanation for the mutant phenotype. Unfortunately, unlike in mice, technologies for conditional knockout of genes in specific cell types are not yet available for our model and cell types. However, in the revised manuscript we will add a supplementary figure describing the distribution of this receptor in other tissues.

It also is not clear whether the phenotypes of the fish reflect perturbations in pituitary development vs. a loss of CCK receptor function in the pituitary later in life. Ideally, the authors would attempt to block CCK signaling in adult fish that develop normally. For example, if CCK receptor antagonists are available, they could be used to treat fish and see whether and how this affects FSH vs. LH secretion.

While the observed gonadal phenotype of the KO (sex inversion) should have a developmental origin since it requires a long time to manifest, the effect of the KO on FSH and LH cells is probably more acute.

In the Discussion, the authors suggest that CCK, as a satiety factor, may provide a link between metabolism and reproduction. This is an interesting idea, but it is not supported by the data presented. That is, none of the results shown link metabolic state to CCK regulation of FSH and fertility. Absent such data, the lengthy Discussion of the link is speculative and not fully merited.

In the revised manuscript, we will address this comment by either providing data to link cck with metabolic status or tuning down the Discussion of this topic.

Also in the Discussion, the authors argue that "CCK directly controls FSH cells by innervating the pituitary gland and binding to specific receptors that are particularly abundant in FSH gonadotrophs." However, their imaging does not demonstrate innervation of FSH cells by CCK terminals (e.g., at the EM level).

Innervation of the fish pituitary does not imply a synaptic-like connection between axon terminals and endocrine cells. In fact, such connections are extremely rare, and their functionality is unclear. Instead, the mode of regulation between hypothalamic terminals and endocrine cells in the fish pituitary is more similar to "volume transmission" in the CNS, i.e. peptides are released into the tissue and carried to their endocrine cell targets by the circulation or via diffusion.

Moreover, they have not demonstrated the binding of CCK to these cells. Indeed, no CCK receptor protein data are shown.

Our revised manuscript will include detailed experiments showing the activation of the receptor by its ligand. Unfortunately, no antibody is available against this fish- specific receptor (one of the caveats of working with fish models); therefore, we cannot present receptor protein data.

The calcium responses of FSH cells to exogenous CCK certainly suggest the presence of functional CCK receptors therein; but, the nature of the preparations (with all pituitary cell types present) does not demonstrate that CCK is acting directly in these cells.

We agree with the reviewer that there are some disadvantages in choosing to work with a whole-tissue preparation. However, we believe that the advantages of working in a more physiological context far outweigh the drawbacks as it reflects the natural dynamics more precisely. Since our transcriptome data as well as our ISH staining, show that the CCK receptor is exclusively expressed on FSH cells, it is improbable that the observed calcium response is mediated via a different pituitary cell type.

Indeed, the asynchrony in responses of individual FSH cells to CCK (Figure 4) suggests that not all cells may be activated in the same way. Contrast the response of LH cells to GnRH, where the onset of calcium signaling is similar across cells (Figure 3).

The difference between the synchronization levels of LH and FSH cells activity stems from the gap-junction mediated coupling between LH cells that does not exist between FSH cells (Golan et al 2016, DOI: 10.1038/srep23777). Therefore, the onset of calcium response in FSH cells is dependent on the irregular diffusion rate of the peptide within the preparation, whereas the tight homotypic coupling between LH cells generates a strong and synchronized calcium rise that propagates quickly throughout the entire population; we will make sure this is clear in the final revision.

Finally, as the authors note in the Discussion, the data presented do not enable them to conclude that the endogenous CCK regulating FSH (assuming it does) is from the brain as opposed to other sources (e.g., the gut).

We agree with the reviewer that, for now, we are unable to determine whether hypothalamic or peripheral CCK are the main drivers of FSH cells. While the strong innervation of the gland by CCK-secreting hypothalamic neurons strengthens the notion of a hypothalamic-releasing hormone and also fits with the dogma of the neural control of the pituitary gland in fish (Ball, 1981; doi: 10.1016/0016-6480(81)90243-4.), more experiments are required to resolve this question.

Reviewer #2 (Public Review):

Summary:

This manuscript builds on previous work suggesting that the CCK peptide is the releasing hormone for FSH in fishes, which is different than that observed in mammals where both LH and FSH release are under the control of GnRH. Based on data using calcium imaging as a readout for stimulation of the gonadotrophs, the researchers present data supporting the hypothesis that CCK stimulates FSH- containing cells in the pituitary. In contrast, LH-containing cells show a weak and variable response to CCK but are highly responsive to GnRH. Data are presented that support the role of CCK in the release of FSH. Researchers also state that functional overlap exists in the potency of GnRH to activate FSH cells, thus the two signalling pathways are not separate.

The results are of interest to the field because for many years the assumption has been that fishes use the same signalling mechanism. These data present an intriguing variation where a hormone involved in satiation acts in the control of reproduction.

Strengths:

The strengths of the manuscript are that researchers have shed light on different pathways controlling reproduction in fishes.

Weaknesses:

Weaknesses are that it is not clear if multiple ligand/receptors are involved (more than one CCK and more than one receptor?). The imaging of the CCK terminals and CCK receptors needs to be reinforced.

Reviewer consultation summary:

  • The data presented establish sufficiency, but not necessity of CCK in FSH regulation. The paper did not show that CCK endogenously regulates FSH in fish. This has not been established yet.

This is a very important comment, also raised by reviewer 1. To avoid repetition, please see our detailed response to the comment above.

  • The paper presents the pharmacological effects of CCK on ex vivo preparations but does not establish the in vivo physiological function of the peptide. The current evidence for a novel physiological regulatory mechanism is incomplete and would require further physiological experiments. These could include the use of a CCK receptor antagonist in adult fish to see the effects on FSH and LH release, the generation of a CCK knockout, or cell-specific genetic manipulations.

As detailed in the responses to the first reviewer,we cannot conduct conditional, cell- specific gene knockout in our model.

  • Zebrafish have two CCK ligands: ccka, cckb and also multiple receptors: cckar, cckbra and cckbrb. There is ambiguity about which CCK receptor and ligand are expressed and which gene was knocked out.

In the revised manuscript, we will clarify which of the receptors are expressed and which receptor is targeted. We will also provide data showing the specificity of the receptors (both WT and mutant) to the ligands.

  • Blocking CCK action in fish (with receptor KO) affects FSH and LH. Therefore, the work did not demonstrate a selective role for CCK in FSH regulation in vivo and any claims to have discovered FSHRH need to be more conservative.

We agree with the reviewer that the overlap in the effect of CCK measured in the calcium activation of cells and in the KO model does not allow us to conclude selectivity. In this context, it is crucial to highlight that CCK-R exhibits high expression on FSH cells but not on LH cells. Therefore, the effect of CCK on LH cells is likely paracrine rather than solely endocrine. We will tone down our claims of selectivity in the revised manuscript.

  • The labelling of the terminals with anti-CCK looks a lot like the background and the authors did not show a specificity control (e.g. anti-CCK antibody pre-absorbed with the peptide or anti-CCK in morphant/KO animals).

We will update the colors of the image for better clarity. Also, The same antibody had been previously used to mark CCK-positive cells in the gut of the red drum fish (K.A. Webb, Jr. 2010; DOI: https://doi.org/10.1016/j.ygcen.2009.10.010), where a control (pre-absorbed with the peptide) experiment had been conducted.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation